1
|
Kong H, Chen X, Lee W, Xie X, Tao Y, Li M. Dual-color fluorescence detection of tumor-derived extracellular vesicles using a specific and serum-stable membrane-fusion approach. Biosens Bioelectron 2025; 278:117302. [PMID: 40101657 DOI: 10.1016/j.bios.2025.117302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/05/2025] [Accepted: 02/21/2025] [Indexed: 03/20/2025]
Abstract
Tumor-derived extracellular vesicles (tEVs), which are essential mediators for cell-to-cell communication during tumorigenesis and tumor development, have demonstrated significant diagnostic potential in cancer liquid biopsy, particularly through biomarkers like membrane proteins and inner microRNAs. However, traditional detection methods such as ELISA and qRT-PCR encounter challenges with low sensitivity and specificity, complex procedures, and high costs. Although emerging biosensors have been developed, these methods are limited to detecting a single type of tEV biomarker, which may result in misdiagnoses due to false-positive or false-negative signals. Herein, we introduce a specific and serum-stable membrane-fusion approach (SSMFA) capable of simultaneously detecting tEV proteins and microRNAs via dual-color fluorescence analysis. In this strategy, the established epithelial cell adhesion molecule (EpCAM) aptamer-modified serum-stable membrane-fusion liposome (AptSMFL) is labeled with fluorescence resonance energy transfer (FRET) dye pairs, which can specifically recognize EpCAM-overexpressed tEVs and induce serum-stable membrane fusion, allowing the quantification of EpCAM protein levels through red fluorescence changes resulting from FRET alterations. Meanwhile, SSMFA facilitates efficient transfection of the CRISPR/Cas13a probe into tEVs to analyze the levels of microRNA-21 (miR-21) in EpCAM-positive tEVs via green fluorescence detection. When tested on serum samples from hepatocellular carcinoma models, the SSMFA exhibited minimal sample volume requirement and rapid assay time (2 h) to effectively achieve accurate quantification of both tEV EpCAM protein and miR-21 levels. Additionally, this dual-biomarker detection method showed a strong correlation with tumor burden and significantly improved cancer diagnostic accuracy (AUC = 0.98), underscoring the potential of SSMFA as a promising tEV-based liquid biopsy assay for cancer diagnosis.
Collapse
Affiliation(s)
- Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaodie Chen
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Weijen Lee
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China.
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-Sen University, Guangzhou 510275, China; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China.
| |
Collapse
|
2
|
Lu C, Kang T, Zhang J, Yang K, Liu Y, Song K, Lin Q, Dixit D, Gimple RC, Zhang Q, Shi Z, Fan X, Wu Q, Li D, Shan D, Gao J, Gu D, You H, Li Y, Yang J, Zhao L, Qiu Z, Yang H, Zhao N, Gao W, Tao W, Lu Y, Chen Y, Ji J, Zhu Z, Kang C, Man J, Agnihotri S, Wang Q, Lin F, Qian X, Mack SC, Hu Z, Li C, Taylor MD, Liu N, Zhang N, Lu M, You Y, Rich JN, Zhang W, Wang X. Combined targeting of glioblastoma stem cells of different cellular states disrupts malignant progression. Nat Commun 2025; 16:2974. [PMID: 40140646 PMCID: PMC11947120 DOI: 10.1038/s41467-025-58366-5] [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: 03/26/2024] [Accepted: 03/19/2025] [Indexed: 03/28/2025] Open
Abstract
Glioblastoma (GBM) is the most lethal primary brain tumor with intra-tumoral hierarchy of glioblastoma stem cells (GSCs). The heterogeneity of GSCs within GBM inevitably leads to treatment resistance and tumor recurrence. Molecular mechanisms of different cellular state GSCs remain unclear. Here, we find that classical (CL) and mesenchymal (MES) GSCs are enriched in reactive immune region and high CL-MES signature informs poor prognosis in GBM. Through integrated analyses of GSCs RNA sequencing and single-cell RNA sequencing datasets, we identify specific GSCs targets, including MEOX2 for the CL GSCs and SRGN for the MES GSCs. MEOX2-NOTCH and SRGN-NFκB axes play important roles in promoting proliferation and maintaining stemness and subtype signatures of CL and MES GSCs, respectively. In the tumor microenvironment, MEOX2 and SRGN mediate the resistance of CL and MES GSCs to macrophage phagocytosis. Using genetic and pharmacologic approaches, we identify FDA-approved drugs targeting MEOX2 and SRGN. Combined CL and MES GSCs targeting demonstrates enhanced efficacy, both in vitro and in vivo. Our results highlighted a therapeutic strategy for the elimination of heterogeneous GSCs populations through combinatorial targeting of MEOX2 and SRGN in GSCs.
Collapse
Affiliation(s)
- Chenfei Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tao Kang
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Yang Liu
- Department of Pharmacology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Kefan Song
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiankun Lin
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Deobrat Dixit
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Ryan C Gimple
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Qian Zhang
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhumei Shi
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Fan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiulian Wu
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Daqi Li
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danyang Shan
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiancheng Gao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danling Gu
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao You
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yangqing Li
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junlei Yang
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Linjie Zhao
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Zhixin Qiu
- Department of Anesthesiology, Zhongshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ningwei Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Gao
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiwei Tao
- College of Biomedicine and Health & College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yingmei Lu
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Chen
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Ji
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhe Zhu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Chunsheng Kang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianghong Man
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Sameer Agnihotri
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Qianghu Wang
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fan Lin
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xu Qian
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Stephen C Mack
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhibin Hu
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chaojun Li
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Michael D Taylor
- Department of Pediatrics- Hematology/Oncology and Neurosurgery, Texas Children's Cancer Center, Hematology-Oncology Section, Baylor College of Medicine, Houston, Texas, USA
| | - Ning Liu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ming Lu
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jeremy N Rich
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xiuxing Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China.
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
- Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
3
|
Rahimi-Farsi N, Bostanian F, Shahbazi T, Shamsinejad FS, Bolideei M, Mohseni P, Zangooie A, Boustani F, Shoorei H. Novel oncogenes and tumor suppressor genes in Hepatocellular Carcinoma: Carcinogenesis, progression, and therapeutic targets. Gene 2025; 941:149229. [PMID: 39800198 DOI: 10.1016/j.gene.2025.149229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 10/21/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Hepatocellular carcinoma (HCC) is the primary malignancy affecting the liver and the leading cause of mortality among individuals with cirrhosis. This complex disease is associated with various risk factors, including environmental, pathological, and genetic influences, which dysregulate gene expression crucial for the cell cycle and cellular/molecular pathways. The disruption of the balance between tumor suppressors and proto-oncogenes amplifies the pathogenic cascade. Given its predilection for diseased or cirrhotic livers and late-stage diagnosis, HCC prognosis is typically poor. Current therapies offer limited benefits, with conventional non-specific cytotoxic agents exhibiting suboptimal efficacy. However, molecularly targeted therapies have emerged as a promising avenue, leveraging the strategic inhibition of carcinogenic molecules to provide heightened specificity and potency compared to cytotoxic chemotherapy. Several clinical trials have demonstrated promising outcomes in advanced HCC with targeted pharmacotherapies. Many genes have been implicated in HCC pathogenesis, underscoring the need to elucidate their molecular functions and roles. This has profound implications for early HCC prognostication via biomarkers and for identifying therapeutic targets to impede neoplastic progression. Notably, evidence highlights the pivotal roles of oncogenes and tumor suppressors in HCC pathophysiology. This discourse examines the potential involvement of ABL1, Annexins, FAK, FOX, and KIF as candidate oncogenes, contrasted with SORBS2, HPCAL1, PCDH10, PLAC8, and CXXC5 as plausible tumor suppressors. Their signaling cascades and relevance to HCC prognosis and progression are delineated to identify targets for improving HCC diagnosis, prognostication, and therapy.
Collapse
Affiliation(s)
| | | | - Taha Shahbazi
- Neurosurgery Research Group (NRG), Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mansoor Bolideei
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Parvin Mohseni
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Alireza Zangooie
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran; Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farnaz Boustani
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran; Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
4
|
Yan B, Lu Q, Gao T, Xiao K, Zong Q, Lv H, Lv G, Wang L, Liu C, Yang W, Jiang G. CD146 regulates the stemness and chemoresistance of hepatocellular carcinoma via JAG2-NOTCH signaling. Cell Death Dis 2025; 16:150. [PMID: 40032820 PMCID: PMC11876685 DOI: 10.1038/s41419-025-07470-x] [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: 08/28/2024] [Revised: 01/29/2025] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
CD146 plays a key role in cancer progression and metastasis. Cancer stem cells (CSCs) are responsible for tumor initiation, drug resistance, metastasis, and recurrence. In this study, we explored the role of CD146 in the regulation of liver CSCs. Here, we demonstrated that CD146 was highly expressed in liver CSCs. CD146 overexpression promoted the self-renewal ability and chemoresistance of Hepatocellular Carcinoma (HCC) cells in vitro and tumorigenicity in vivo. Inversely, knockdown of CD146 restrained these abilities. Mechanistically, CD146 activated the NF-κB signaling to up-regulate JAG2 expression and activated the Notch signaling, which resulted in increased stemness of HCC. Furthermore, JAG2 overexpression restored the Notch signaling activity, the stemness, and chemotherapeutic resistance caused by CD146 knockdown. These results demonstrated that CD146 positively regulates HCC stemness by activating the JAG2-NOTCH signaling. Combined targeting of CD146 and JAG2 may represent a novel therapeutic strategy for HCC treatment.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/drug therapy
- Humans
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/drug therapy
- Jagged-2 Protein/metabolism
- Jagged-2 Protein/genetics
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/drug effects
- Signal Transduction
- Receptors, Notch/metabolism
- Drug Resistance, Neoplasm/genetics
- Animals
- CD146 Antigen/metabolism
- CD146 Antigen/genetics
- Mice
- Cell Line, Tumor
- Mice, Nude
- Gene Expression Regulation, Neoplastic
- Mice, Inbred BALB C
- Male
Collapse
Affiliation(s)
- Bing Yan
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
- Department of General Surgery, Pingxiang People's Hospital, Pingxiang, 337000, China
| | - QiuYu Lu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - TianMing Gao
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - KunQing Xiao
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - QianNi Zong
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - HongWei Lv
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - GuiShuai Lv
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Liang Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - ChunYing Liu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China.
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China.
- Shanghai Key Laboratory of Hepatobiliary Tumor Biology, Shanghai, 200438, China.
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| | - Wen Yang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Naval Medical University (Second Military Medical University), Shanghai, 200438, China.
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China.
- Shanghai Key Laboratory of Hepatobiliary Tumor Biology, Shanghai, 200438, China.
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| | - GuoQing Jiang
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China.
| |
Collapse
|
5
|
Wang T, Li W, Wu Y, You L, Zheng C, Zhang J, Qu L, Sun X. Construction of a prognostic model based on disulfidptosis-related genes and identification of CCNA2 as a novel biomarker for hepatocellular carcinoma. Biol Direct 2024; 19:128. [PMID: 39695705 DOI: 10.1186/s13062-024-00569-9] [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: 09/03/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Disulfidptosis, identified as an innovative form of cellular death subsequent to cuproptosis, is currently under investigation for its mechanisms in oncological contexts. In-depth analyses exploring the relationship between disulfidptosis-related genes (DRGs) and hepatocellular carcinoma (HCC) are currently limited. METHODS Transcriptomic data and clinical information were retrieved from the TCGA and GEO databases (GSE76427 and GSE54236), concentrating on the expression levels of 24 DRGs. Subsequently, multifactor and LASSO regression analyses were utilized to construct the 5-DRG prognostic signature. Immunohistochemistry (IHC) was employed to assess Cyclin A2 (CCNA2) protein expression levels. Quantitative real-time PCR (qRT-PCR) and western blot analyses were conducted to detect transcriptomic and protein expression of CCNA2-targeting short interfering RNA (siRNA). The Cell Counting Kit-8 (CCK-8) assay, EdU staining, and scratch experiments were employed to observe the proliferation and migration of hepatoma cell lines subsequent to CCNA2 inhibition. RESULTS Three HCC patterns were identified, among which pattern B exhibited the the most unfavorable survival outcomes. Five DRGs (STC2, PBK, CCNA2, SERPINE1, and SLC6A1) were involved to establish the 5-DRG prognostic signature. High-risk groups (HRGs) exhibited prolonged survival durations in comparison to low-risk groups (LRGs). Both bioinformatics analyses and experimental methodologies corroborated the association of CCNA2 with poor prognosis in HCC patients. Functional studies elucidated that interference with CCNA2 significantly inhibited proliferation and migration, while simultaneously promoting apoptosis in hepatoma cells and resulting in the downregulation of epithelial-mesenchymal transition (EMT)-related protein markers. CONCLUSIONS The 5-DRG prognostic signature is proficient in predicting clinical outcomes, informing therapeutic strategies, and elucidating the characteristics of the immune microenvironment in HCC patients. Furthermore, this study elucidates the potential of CCNA2 as an innovative biomarker for HCC.
Collapse
Affiliation(s)
- Tao Wang
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wenxuan Li
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuelan Wu
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Liping You
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chao Zheng
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jinghao Zhang
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lihong Qu
- Department of Infectious Diseases, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Xuehua Sun
- Department of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
6
|
Shao G, Wang X, Zheng Y, Ma J, Wang L, Yan Z, Sun Z, Zhang S, Wu H, Lv Y, Huang H, Li J, Zhu T, Yang B, Wang N, Chen T, Guo X, Jin Y, Kang J, Wang H, Cao Y, Fu C. Identification of ANXA1 as a Novel Upstream Negative Regulator of Notch1 Function in AML. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2409726. [PMID: 39447086 DOI: 10.1002/advs.202409726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/06/2024] [Indexed: 10/26/2024]
Abstract
Abnormal Notch1 expression has an important role in tumorigenesis. However, upstream control mechanisms for Notch1 are still insufficiently understood. Acute myeloid leukemia (AML) is one of the most common and lethal blood malignancies with limited possibilities for treatment. Thus, new therapeutic targets are urgently needed to improve current ineffective therapies. Herein, high Annexin A1 (ANXA1) expression is found correlated with hyperproliferation of AML cells, and then ANXA1 is identified as a novel negative regulator of Notch1 function in AML. Mechanistically, ANXA1 directly bound to the intracellular domain of Notch1 (NICD) to target this tumor suppressor for degradation. Furthermore, NICD executed its tumor suppressive function through activation of the p15 promoter. Thus, ablation of the Notch1-p15-mediated tumor suppression by ANXA1 provided a novel mechanism of AML proliferation. In human AML patients, a mutual exclusive relation is discovered between ANXA1 and Notch1/p15, corroborating mechanistic discovery. On the basis of these results, it is reasonably speculated that targeting ANXA1 would provide an effective approach for treatment of AML. In support of this new therapeutic paradigm, provided proof-of-concept data by antagonizing ANXA1 using NICD inhibitory peptides.
Collapse
Affiliation(s)
- Gang Shao
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Department of Oncology, No.903 Hospital of PLA Joint Logistic Support Force, Hangzhou, 310013, China
- College of Life Sciences, China Jiliang University, Hangzhou, 310018, China
| | - Xi Wang
- Department of Oncology, No.903 Hospital of PLA Joint Logistic Support Force, Hangzhou, 310013, China
| | - Yiting Zheng
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Junjie Ma
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Lei Wang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Zhibin Yan
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Zeyu Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shuyuan Zhang
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Hongzhang Wu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Yudie Lv
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- College of Life Sciences, China Jiliang University, Hangzhou, 310018, China
| | - Hemiao Huang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Jianhu Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tianyi Zhu
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Bing Yang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Nanxi Wang
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Tao Chen
- Sartorius (Shanghai) Trading Co., Ltd., Shanghai, 200120, China
| | - Xuancheng Guo
- Hangzhou Acnovia Biotech Co., Ltd., Hangzhou, 310018, China
| | - Yuanting Jin
- College of Life Sciences, China Jiliang University, Hangzhou, 310018, China
| | - Jian Kang
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, 305 Grattan street, Melbourne, Victoria, 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - Huafeng Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, 171 77, Sweden
| | - Caiyun Fu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
7
|
Hu J, Chen NN, Li LG, Yu TT, Qin Y, Peng XC, Li HT, Li XY, Ma TQ, Lu YH, Han N, Xu Z, Hui YJ, Li TF. Cepharanthine-mediated endoplasmic reticulum stress inhibits Notch1 via binding GRP78 for suppressing hepatocellular carcinoma metastasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156162. [PMID: 39427519 DOI: 10.1016/j.phymed.2024.156162] [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: 12/02/2023] [Revised: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND The metastasis of hepatocellular carcinoma (HCC) leads to a poor prognosis, wherein the activation of Notch1 is an essential contributor. Cepharanthine (Cep) has been identified for its effective antiviral function and versatile intracellular targets. Our previous study has only reported the anti-cancer efficacy of Cep in lung cancer, without an in-depth exploration. Herein, the present study aims to investigate the anti-metastasis effect in HCC, the target involved, and the molecular mechanism of Cep. METHODS Stable over-expression of Notch1-N1ICD yielded C5WN1 cells compared with C5WBF344 cells. The C5WN1 cells and C5WN1 cell-bearing mice were applied as the HCC model. The bioinformatics analysis, RNA sequencing, molecular docking, cellular thermal shift assay (CETSA), drug affinity responsive target stability (DARTS), microscale thermophoresis (MST), and transient knockdown techniques were carried out to identify the underlying target. The apoptosis assay, immunofluorescent staining, qRT-PCR, Western blots, Elisa, flow cytometry, migration and scratching experiments, Transmission electron microscopy (TEM), laser scanning confocal microscopy (LSCM), micro-computed tomography (micro-CT), and histopathological experiments were conducted to assay the anti-HCC efficacy, functions, and mechanism. RESULTS Notch1 had an increased expression in HCC and contributed to metastasis thereupon. Surprisingly, Cep (2 μg/ml in vitro, 5 mg kg-1in vivo) presented potent Notch1 signaling pathway inhibitory effect and anti-metastasis efficacy in C5WN1 cells and in situ mice models as evidenced by reduced Notch1/MMP-2/MMP-9 expression, TGF-β release, decreased cell migration, diminished pulmonary metastases, and prolonged survival. RNA sequencing showed that the differential gene of Cep-treated HCC cells was positioned in the endoplasmic reticulum (ER). Molecular docking, CETSA, DARTS, and MST further identified that the possible target of Cep was GRP78, which was distributed in the ER. As expected, Cep (2 μg/ml) up-regulated the critical molecules of ER stress such as GRP78, induced β-amyloid accumulation, and promoted calcium burst in HCC. In contrast, suppression of GRP78 attenuated Cep-induced ER stress. Furthermore, inhibition of ER stress abated Cep-induced Notch1 inactivation and HCC cells' migration. CONCLUSIONS Taken together, the present study finds that Cep possesses excellent anti-metastasis of HCC, wherein the GRP78 could be directly bound and activated by Cep, leading to ER stress and Notch1 blockage. This study reveals for the first time the effect, critical target, and mechanism of the Cep-mediated anti-cancer effect, providing novel insights into the molecular target therapy by phytomedicine.
Collapse
Affiliation(s)
- Jun Hu
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Nan-Nan Chen
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Liu-Gen Li
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Ting-Ting Yu
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Yufei Qin
- Digestive Medicine Center, the Seventh Affiliated Hospital of Sun Yat-sen University, Zhenyuan road No 628, Shenzhen 442000, Guangdong, China
| | - Xing-Chun Peng
- Department of Pathology, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen 518118, Guangzhou Province, China
| | - Hai-Tao Li
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Xian-Yu Li
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Tian-Qi Ma
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Yao-Hua Lu
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Ning Han
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Zhijie Xu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Yuan-Jian Hui
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin south road No 32, Shiyan 442000, Hubei, China.
| | - Tong-Fei Li
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China.
| |
Collapse
|
8
|
Dong H, Wen C, He L, Zhang J, Xiang N, Liang L, Hu L, Li W, Liu J, Shi M, Hu Y, Chen S, Liu H, Yang X. Nilotinib boosts the efficacy of anti-PDL1 therapy in colorectal cancer by restoring the expression of MHC-I. J Transl Med 2024; 22:769. [PMID: 39143573 PMCID: PMC11325812 DOI: 10.1186/s12967-024-05572-2] [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: 05/20/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Although immune checkpoint inhibitors (ICIs) have revolutionized the landscape of cancer treatment, only a minority of colorectal cancer (CRC) patients respond to them. Enhancing tumor immunogenicity by increasing major histocompatibility complex I (MHC-I) surface expression is a promising strategy to boost the antitumor efficacy of ICIs. METHODS Dual luciferase reporter assays were performed to find drug candidates that can increase MHC-I expression. The effect of nilotinib on MHC-I expression was verified by dual luciferase reporter assays, qRT-PCR, flow cytometry and western blotting. The biological functions of nilotinib were evaluated through a series of in vitro and in vivo experiments. Using RNA-seq analysis, immunofluorescence assays, western blotting, flow cytometry, rescue experiments and microarray chip assays, the underlying molecular mechanisms were investigated. RESULTS Nilotinib induces MHC-I expression in CRC cells, enhances CD8+ T-cell cytotoxicity and subsequently enhances the antitumor effects of anti-PDL1 in both microsatellite instability and microsatellite stable models. Mechanistically, nilotinib promotes MHC-I mRNA expression via the cGAS-STING-NF-κB pathway and reduces MHC-I degradation by suppressing PCSK9 expression in CRC cells. PCSK9 may serve as a potential therapeutic target for CRC, with nilotinib potentially targeting PCSK9 to exert anti-CRC effects. CONCLUSION This study reveals a previously unknown role of nilotinib in antitumor immunity by inducing MHC-I expression in CRC cells. Our findings suggest that combining nilotinib with anti-PDL1 therapy may be an effective strategy for the treatment of CRC.
Collapse
Affiliation(s)
- Haiyan Dong
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Chuangyu Wen
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, 523059, Guangdong, China.
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, 60637, USA.
| | - Lu He
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Department of Neurology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Jingdan Zhang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Nanlin Xiang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Liumei Liang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Limei Hu
- Department of Clinical Laboratory Medicine, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Weiqian Li
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Jiaqi Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Mengchen Shi
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Yijia Hu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Siyu Chen
- Guangdong Laboratory, GuangdongKey Laboratory Animal Lab, Animals Monitoring Institute, Guangzhou, 510633, Guangdong, China
| | - Huanliang Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China.
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China.
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China.
| | - Xiangling Yang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China.
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China.
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China.
| |
Collapse
|
9
|
Eshaq AM, Flanagan TW, Hassan SY, Al Asheikh SA, Al-Amoudi WA, Santourlidis S, Hassan SL, Alamodi MO, Bendhack ML, Alamodi MO, Haikel Y, Megahed M, Hassan M. Non-Receptor Tyrosine Kinases: Their Structure and Mechanistic Role in Tumor Progression and Resistance. Cancers (Basel) 2024; 16:2754. [PMID: 39123481 PMCID: PMC11311543 DOI: 10.3390/cancers16152754] [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: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Protein tyrosine kinases (PTKs) function as key molecules in the signaling pathways in addition to their impact as a therapeutic target for the treatment of many human diseases, including cancer. PTKs are characterized by their ability to phosphorylate serine, threonine, or tyrosine residues and can thereby rapidly and reversibly alter the function of their protein substrates in the form of significant changes in protein confirmation and affinity for their interaction with protein partners to drive cellular functions under normal and pathological conditions. PTKs are classified into two groups: one of which represents tyrosine kinases, while the other one includes the members of the serine/threonine kinases. The group of tyrosine kinases is subdivided into subgroups: one of them includes the member of receptor tyrosine kinases (RTKs), while the other subgroup includes the member of non-receptor tyrosine kinases (NRTKs). Both these kinase groups function as an "on" or "off" switch in many cellular functions. NRTKs are enzymes which are overexpressed and activated in many cancer types and regulate variable cellular functions in response to extracellular signaling-dependent mechanisms. NRTK-mediated different cellular functions are regulated by kinase-dependent and kinase-independent mechanisms either in the cytoplasm or in the nucleus. Thus, targeting NRTKs is of great interest to improve the treatment strategy of different tumor types. This review deals with the structure and mechanistic role of NRTKs in tumor progression and resistance and their importance as therapeutic targets in tumor therapy.
Collapse
Affiliation(s)
- Abdulaziz M. Eshaq
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA;
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Sofie-Yasmin Hassan
- Department of Pharmacy, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sara A. Al Asheikh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Waleed A. Al-Amoudi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Simeon Santourlidis
- Institute of Cell Therapeutics and Diagnostics, University Medical Center of Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sarah-Lilly Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Maryam O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Marcelo L. Bendhack
- Department of Urology, Red Cross University Hospital, Positivo University, Rua Mauá 1111, Curitiba 80030-200, Brazil;
| | - Mohammed O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| | - Mohamed Hassan
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
10
|
Zhao N, Ni C, Zhang D, Che N, Li Y, Wang X. Identification of a vascular invasion-related signature based on lncRNA pairs for predicting prognosis in hepatocellular carcinoma. BMC Gastroenterol 2024; 24:33. [PMID: 38221614 PMCID: PMC10788995 DOI: 10.1186/s12876-023-03118-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 12/31/2023] [Indexed: 01/16/2024] Open
Abstract
OBJECTIVES Most signatures are constructed on the basis of RNA or protein expression levels. The value of vascular invasion-related signatures based on lncRNA pairs, regardless of their specific expression level in hepatocellular carcinoma (HCC), is not yet clear. METHODS Vascular invasion-related differentially expressed lncRNA (DElncRNA) pairs were identified with a two-lncRNA combination strategy by using a novel modeling algorithm. Based on the optimal cutoff value of the ROC curve, patients with HCC were classified into high- and low-risk subgroups. We used KM survival analysis to evaluate the overall survival rate of patients in the high- and low-risk subgroups. The independent indicators of survival were identified using univariate and multivariate Cox analyses. RESULTS Five pairs of vascular invasion-related DElncRNAs were selected to develop a predictive model for HCC. High-risk subgroups were closely associated with aggressive clinicopathological characteristics and genes, chemotherapeutic sensitivity, and highly expressed immune checkpoint inhibitors. CONCLUSIONS We identified a signature composed of 5 pairs of vascular invasion-related lncRNAs that does not require absolute expression levels of lncRNAs and shows promising clinical predictive value for HCC prognosis. This predictive model provides deep insight into the value of vascular invasion-related lncRNAs in prognosis.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Pathology, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China.
- Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, 300052, China.
| | - Chunsheng Ni
- Department of Pathology, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
- Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
- Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Na Che
- Department of Pathology, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
- Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Yanlei Li
- Department of Pathology, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
- Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Xiao Wang
- Department of Pathology, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| |
Collapse
|
11
|
Hou W, Gad SA, Ding X, Dhanarajan A, Qiu W. Focal adhesion kinase confers lenvatinib resistance in hepatocellular carcinoma via the regulation of lysine-deficient kinase 1. Mol Carcinog 2024; 63:173-189. [PMID: 37787401 PMCID: PMC10842616 DOI: 10.1002/mc.23644] [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: 05/01/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 10/04/2023]
Abstract
Lenvatinib is a clinically effective multikinase inhibitor approved for first-line therapy of advanced hepatocellular carcinoma (HCC). Although resistance against lenvatinib often emerges and limits its antitumor activity, the underlying molecular mechanisms involved in endogenous and acquired resistance remain elusive. In this study, we identified focal adhesion kinase (FAK) as a critical contributor to lenvatinib resistance in HCC. The elevated expression and phosphorylation of FAK were observed in both acquired and endogenous lenvatinib-resistant (LR) HCC cells. Furthermore, inhibition of FAK reversed lenvatinib resistance in vitro and in vivo. Mechanistically, FAK promoted lenvatinib resistance through regulating lysine-deficient kinase 1 (WNK1). Phosphorylation of WNK1 was significantly increased in LR-HCC cells. Further, WNK1 inhibitor WNK463 resensitized either established or endogenous LR-HCC cells to lenvatinib treatment. In addition, overexpression of WNK1 desensitized parental HCC cells to lenvatinib treatment. Conclusively, our results establish a crucial role and novel mechanism of FAK in lenvatinib resistance and suggest that targeting the FAK/WNK1 axis is a promising therapeutic strategy in HCC patients showing lenvatinib resistance.
Collapse
Affiliation(s)
- Wei Hou
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, 2160 South 1 Avenue., Maywood, IL 60153, USA
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1 Avenue., Maywood, IL 60153, USA
| | - Shaimaa A. Gad
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, 2160 South 1 Avenue., Maywood, IL 60153, USA
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1 Avenue., Maywood, IL 60153, USA
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Center, Egypt
| | - Xianzhong Ding
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, 2160 South 1 Avenue., Maywood, IL 60153, USA
| | - Asha Dhanarajan
- Department of Medicine, Loyola University Chicago Stritch School of Medicine, 2160 South 1 Avenue., Maywood, IL 60153, USA
| | - Wei Qiu
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, 2160 South 1 Avenue., Maywood, IL 60153, USA
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1 Avenue., Maywood, IL 60153, USA
| |
Collapse
|
12
|
Ursic-Bedoya J, Gregoire D. Hydrodynamic Transfection of Hepatocytes for the Study of Hepatocellular Carcinogenesis. Methods Mol Biol 2024; 2769:77-85. [PMID: 38315390 DOI: 10.1007/978-1-0716-3694-7_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Hydrodynamic tail vein injection (HTVi), also called hydrodynamic gene transfer (HGT), is attracting increasing interest for modeling hepatic carcinogenesis. This highly versatile approach reproducibly provides efficient in vivo transfection of hepatocytes with naked DNA. Here, we give an in-depth description of the injection procedure, which is key for the success of the method. HTVi requires the injection of a large volume of a solution containing plasmids into the tail vein of the mouse. The transient right heart overload created by the injection forces the blood to flow back into the hepatic veins, enlarging the endothelial fenestrae and permeabilizing a fraction of hepatocytes for a few seconds. This results in the uptake of plasmids by the permeabilized hepatocytes, giving rise to their in vivo transfection. Including the Sleeping Beauty transposon system among the injected plasmids leads to the stable transfection of a subset of hepatocytes. HTVi is a powerful technique which enables numerous applications in liver cancer biology, such as a study of oncogene cooperation, of tumor heterogeneity, and interaction with the tumor microenvironment.
Collapse
Affiliation(s)
- José Ursic-Bedoya
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
- Department of Hepatogastroenterology, Hepatology and Liver Transplantation Unit, Saint Eloi Hospital, University of Montpellier, Montpellier, France
| | - Damien Gregoire
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
13
|
Malnassy G, Keating CR, Gad S, Bridgeman B, Perera A, Hou W, Cotler SJ, Ding X, Choudhry M, Sun Z, Koleske AJ, Qiu W. Inhibition of Abelson Tyrosine-Protein Kinase 2 Suppresses the Development of Alcohol-Associated Liver Disease by Decreasing PPARgamma Expression. Cell Mol Gastroenterol Hepatol 2023; 16:685-709. [PMID: 37460041 PMCID: PMC10520367 DOI: 10.1016/j.jcmgh.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/07/2023]
Abstract
BACKGROUND & AIMS Alcohol-associated liver disease (ALD) represents a spectrum of alcohol use-related liver diseases. Outside of alcohol abstinence, there are currently no Food and Drug Administration-approved treatments for advanced ALD, necessitating a greater understanding of ALD pathogenesis and potential molecular targets for therapeutic intervention. The ABL-family proteins, including ABL1 and ABL2, are non-receptor tyrosine kinases that participate in a diverse set of cellular functions. We investigated the role of the ABL kinases in alcohol-associated liver disease. METHODS We used samples from patients with ALD compared with healthy controls to elucidate a clinical phenotype. We established strains of liver-specific Abl1 and Abl2 knockout mice and subjected them to the National Institute on Alcohol Abuse and Alcoholism acute-on-chronic alcohol feeding regimen. Murine samples were subjected to RNA sequencing, AST, Oil Red O staining, H&E staining, Western blotting, and quantitative polymerase chain reaction to assess phenotypic changes after alcohol feeding. In vitro modeling in HepG2 cells as well as primary hepatocytes from C57BL6/J mice was used to establish this mechanistic link of ALD pathogenesis. RESULTS We demonstrate that the ABL kinases are highly activated in ALD patient liver samples as well as in liver tissues from mice subjected to an alcohol feeding regimen. We found that the liver-specific knockout of Abl2, but not Abl1, attenuated alcohol-induced steatosis, liver injury, and inflammation. Subsequent RNA sequencing and gene set enrichment analyses of mouse liver tissues revealed that relative to wild-type alcohol-fed mice, Abl2 knockout alcohol-fed mice exhibited numerous pathway changes, including significantly decreased peroxisome proliferator activated receptor (PPAR) signaling. Further examination revealed that PPARγ, a previously identified regulator of ALD pathogenesis, was induced upon alcohol feeding in wild-type mice, but not in Abl2 knockout mice. In vitro analyses revealed that shRNA-mediated knockdown of ABL2 abolished the alcohol-induced accumulation of PPARγ as well as subsequent lipid accumulation. Conversely, forced overexpression of ABL2 resulted in increased PPARγ protein expression. Furthermore, we demonstrated that the regulation of hypoxia inducible factor 1 subunit alpha (HIF1α) by ABL2 is required for alcohol-induced PPARγ expression. Furthermore, treatment with ABL kinase inhibitors attenuated alcohol-induced PPARγ expression, lipid droplet formation, and liver injury. CONCLUSIONS On the basis of our current evidence, we propose that alcohol-induced ABL2 activation promotes ALD through increasing HIF1α and the subsequent PPARγ expression, and ABL2 inhibition may serve as a promising target for the treatment of ALD.
Collapse
Affiliation(s)
- Greg Malnassy
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois; Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Claudia R Keating
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois; Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Shaimaa Gad
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois; Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois; Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Bryan Bridgeman
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois; Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Aldeb Perera
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois; Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Wei Hou
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois; Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Scott J Cotler
- Department of Medicine, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Xianzhong Ding
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Mashkoor Choudhry
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Zhaoli Sun
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut
| | - Wei Qiu
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois; Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois.
| |
Collapse
|
14
|
Zhou Y, Miao Y, Huang Q, Shi W, Xie J, Lin J, Huang P, Yue C, Qin Y, Yu X, Wang H, Qin L, Chen J. A redox-responsive self-assembling COA-4-arm PEG prodrug nanosystem for dual drug delivery suppresses cancer metastasis and drug resistance by downregulating hsp90 expression. Acta Pharm Sin B 2023; 13:3153-3167. [PMID: 37521875 PMCID: PMC10372829 DOI: 10.1016/j.apsb.2022.11.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/09/2022] [Accepted: 11/04/2022] [Indexed: 11/26/2022] Open
Abstract
Metastasis and resistance are main causes to affect the outcome of the current anticancer therapies. Heat shock protein 90 (Hsp90) as an ATP-dependent molecular chaperone takes important role in the tumor metastasis and resistance. Targeting Hsp90 and downregulating its expression show promising in inhibiting tumor metastasis and resistance. In this study, a redox-responsive dual-drug nanocarrier was constructed for the effective delivery of a commonly used chemotherapeutic drug PTX, and a COA-modified 4-arm PEG polymer (4PSC) was synthesized. COA, an active component in oleanolic acid that exerts strong antitumor activity by downregulating Hsp90 expression, was used as a structural and functional element to endow 4PSC with redox responsiveness and Hsp90 inhibitory activity. Our results showed that 4PSC/PTX nanomicelles efficiently delivered PTX and COA to tumor locations without inducing systemic toxicity. By blocking the Hsp90 signaling pathway, 4PSC significantly enhanced the antitumor effect of PTX, inhibiting tumor proliferation and invasiveness as well as chemotherapy-induced resistance in vitro. Remarkable results were further confirmed in vivo with two preclinical tumor models. These findings demonstrate that the COA-modified 4PSC drug delivery nanosystem provides a potential platform for enhancing the efficacy of chemotherapies.
Collapse
Affiliation(s)
- Yi Zhou
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yingling Miao
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Qiudi Huang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Wenwen Shi
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jiacui Xie
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiachang Lin
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Pei Huang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chengfeng Yue
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
- Center of Cancer Research, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Yuan Qin
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiyong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - He Wang
- Center of Cancer Research, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Linghao Qin
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jianhai Chen
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
15
|
Wang G, Zhou Y, Yi B, Long Y, Ma B, Zhang Y. Comprehensive analysis of the prognostic value and biological function of TDG in hepatocellular carcinoma. Cell Cycle 2023; 22:1478-1495. [PMID: 37224078 PMCID: PMC10281473 DOI: 10.1080/15384101.2023.2216501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/30/2023] [Accepted: 04/27/2023] [Indexed: 05/26/2023] Open
Abstract
Epigenetics plays an important role in the malignant progression of tumors, in which DNA methylation can alter genetic performance without altering the DNA sequence. As a key regulator demethylation, thymine-DNA glycosylase (TDG) has been reported to participate in malignant progression of multiple tumors. In this study, we demonstrate that TDG is highly expressed in hepatocellular carcinoma (HCC) and its high expression is closely related to the poor prognosis of patients. Decreasing TDG expression can significantly inhibit the malignant biological behavior of HCC cells. ABL proto-oncogene 1(ABL1) was identified as a downstream gene regulated by TDG demethylation. In addition, TDG can affect the Hippo signaling pathway through ABL1 to regulate HCC cell proliferation, apoptosis, invasion and migration. Overall, our study demonstrated that TDG reduces DNA methylation of ABL1, increases ABL1 protein expression, and affects the Hippo signaling pathway to regulate the malignant progression of HCC.
Collapse
Affiliation(s)
- Guoliang Wang
- Department of Hepatobiliary Surgery, Department of Organ Transplantation, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Yinwen Zhou
- Department of Surgery, Zunyi Medical University, Zunyi, Guizhou, China
| | - Bin Yi
- Department of Hepatobiliary Surgery, Department of Organ Transplantation, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Yanli Long
- Department of Pathology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Bo Ma
- Department of Hepatobiliary Surgery, Department of Organ Transplantation, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Yi Zhang
- Department of Hepatobiliary Surgery, Department of Organ Transplantation, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| |
Collapse
|
16
|
Jiang M, Guo R, Ai Y, Wang G, Tang P, Jia X, He B, Yuan Q, Xie X. Small molecule drugs promote repopulation of transplanted hepatocytes by stimulating cell dedifferentiation. JHEP Rep 2023; 5:100670. [PMID: 36873420 PMCID: PMC9976449 DOI: 10.1016/j.jhepr.2023.100670] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 01/15/2023] Open
Abstract
Background & Aims Hepatocyte transplantation has emerged as a possible treatment option for end-stage liver disease. However, an important obstacle to therapeutic success is the low level of engraftment and proliferation of transplanted hepatocytes, which do not survive long enough to exert therapeutic effects. Thus, we aimed to explore the mechanisms of hepatocyte proliferation in vivo and find a way to promote the growth of transplanted hepatocytes. Methods Hepatocyte transplantation was performed in Fah -/- mice to explore the mechanisms of hepatocyte proliferation in vivo. Guided by in vivo regeneration mechanisms, we identified compounds that promote hepatocyte proliferation in vitro. The in vivo effects of these compounds on transplanted hepatocytes were then evaluated. Results The transplanted mature hepatocytes were found to dedifferentiate into hepatic progenitor cells (HPCs), which proliferate and then convert back to a mature state at the completion of liver repopulation. The combination of two small molecules Y-27632 (Y, ROCK inhibitor) and CHIR99021 (C, Wnt agonist) could convert mouse primary hepatocytes into HPCs, which could be passaged for more than 30 passages in vitro. Moreover, YC could stimulate the proliferation of transplanted hepatocytes in Fah -/- livers by promoting their conversion into HPCs. Netarsudil (N) and LY2090314 (L), two clinically used drugs which target the same pathways as YC, could also promote hepatocyte proliferation in vitro and in vivo, by facilitating HPC conversion. Conclusions Our work suggests drugs promoting hepatocyte dedifferentiation may facilitate the growth of transplanted hepatocytes in vivo and may facilitate the application of hepatocyte therapy. Impact and implications Hepatocyte transplantation may be a treatment option for patients with end-stage liver disease. However, one important obstacle to hepatocyte therapy is the low level of engraftment and proliferation of the transplanted hepatocytes. Herein, we show that small molecule compounds which promote hepatocyte proliferation in vitro by facilitating dedifferentiation, could promote the growth of transplanted hepatocytes in vivo and may facilitate the application of hepatocyte therapy.
Collapse
Key Words
- (i)HPCs, (induced) hepatic progenitor cells
- A, A-83-01
- ALP, alkaline phosphatase
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- C, CHIR99021
- DDC, 3,5-diethoxycarbonyl-1,4-dihydrocollidine
- Dedifferentiation
- HMM, hepatic maturation medium
- Hepatocyte expansion
- Hepatocyte progenitor cells
- Hepatocyte transplantation
- L, LY2090314
- N, netarsudil
- NTBC, 2-(2-nitro-4-trifluoro-methylbenzoyl)-1,3-cyclo-hexanedione
- PHx, partial hepatectomy
- RT-PCR, reverse-transcription PCR
- Small molecule compounds
- Y, Y27632
- iMHs, induced mature hepatocytes
Collapse
Affiliation(s)
- Mengmeng Jiang
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China.,CAS Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Ren Guo
- CAS Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yan Ai
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Gang Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Peilan Tang
- School of Pharmaceutical Science, Nanchang University, Nanchang 330006, PR China
| | - Xiaohui Jia
- CAS Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Bingqing He
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China.,CAS Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Qianting Yuan
- CAS Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xin Xie
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China.,CAS Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
17
|
Tang H, Zhang H, Liu D, Wang Z, Yu D, Fan W, Guo Z, Huang W, Hou S, Zhou Z. Genome-wide association study reveals the genetic determinism of serum biochemical indicators in ducks. BMC Genomics 2022; 23:856. [PMID: 36575369 PMCID: PMC9795613 DOI: 10.1186/s12864-022-09080-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The serum is rich in nutrients and plays an essential role in electrolyte and acid-base balance, maintaining cellular homeostasis. In addition, serum parameters have been commonly used as essential biomarkers for clinical diagnosis. However, little is known about the genetic mechanism of the serum parameters in ducks. RESULTS This study measured 18 serum parameters in 320 samples of the F2 segregating population generated by Mallard × Pekin duck. The phenotypic correlations showed a high correlation between LDH, HBDH, AST, and ALT (0.59-0.99), and higher coefficients were also observed among TP, ALB, HDL-C, and CHO (0.46-0.87). And then, we performed the GWAS to reveal the genetic basis of the 18 serum biochemical parameters in ducks. Fourteen candidate protein-coding genes were identified with enzyme traits (AST, ALP, LDH, HBDH), and 3 protein-coding genes were associated with metabolism and protein-related serum parameters (UA, TG). Moreover, the expression levels of the above candidate protein-coding genes in different stages of breast muscle and different tissues were analyzed. Furthermore, the genes located within the high-LD region (r2 > 0.4 and - log10(P) < 4) neighboring the significant locus also remained. Finally, 86 putative protein-coding genes were used for GO and KEGG enrichment analysis, the enzyme-linked receptor protein signaling pathway and ErbB signaling pathway deserve further focus. CONCLUSIONS The obtained results can contribute to new insights into blood metabolism and provide new genetic biomarkers for application in duck breeding programs.
Collapse
Affiliation(s)
- Hehe Tang
- grid.410727.70000 0001 0526 1937Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - He Zhang
- grid.410727.70000 0001 0526 1937Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Dapeng Liu
- grid.410727.70000 0001 0526 1937Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Zhen Wang
- grid.410727.70000 0001 0526 1937Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Daxin Yu
- grid.410727.70000 0001 0526 1937Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Wenlei Fan
- grid.412608.90000 0000 9526 6338College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, PR China
| | - Zhanbao Guo
- grid.410727.70000 0001 0526 1937Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Wei Huang
- grid.410727.70000 0001 0526 1937Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Shuisheng Hou
- grid.410727.70000 0001 0526 1937Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Zhengkui Zhou
- grid.410727.70000 0001 0526 1937Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| |
Collapse
|
18
|
IL6 supports long-term expansion of hepatocytes in vitro. Nat Commun 2022; 13:7345. [PMID: 36446858 PMCID: PMC9708838 DOI: 10.1038/s41467-022-35167-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022] Open
Abstract
Hepatocytes are very difficult to expand in vitro. A few studies have demonstrated that chemical cocktails with growth factors or Wnt ligands can support long-term expansion of hepatocytes via dedifferentiation. However, the culture conditions are complex, and clonal expansion of hepatic progenitors with full differentiation capacity are rarely reported. Here, we discover IL6, combined with EGF and HGF, promotes long-term expansion (>30 passages in ~150 days with theoretical expansion of ~1035 times) of primary mouse hepatocytes in vitro in simple 2D culture, by converting hepatocytes into induced hepatic progenitor cells (iHPCs), which maintain the capacity of differentiation into hepatocytes. IL6 also supports the establishment of single hepatocyte-derived iHPC clones. The summation of the downstream STAT3, ERK and AKT pathways induces a number of transcription factors which support rapid growth. This physiological and simple way may provide ideas for culturing previously difficult-to-culture cell types and support their future applications.
Collapse
|
19
|
Liu L, Liu Z, Gao J, Liu X, Weng S, Guo C, Hu B, Wang Z, Zhang J, Shi J, Guo W, Zhang S. CD8+ T cell trajectory subtypes decode tumor heterogeneity and provide treatment recommendations for hepatocellular carcinoma. Front Immunol 2022; 13:964190. [PMID: 35967384 PMCID: PMC9363578 DOI: 10.3389/fimmu.2022.964190] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Mounting evidence has revealed that the interactions and dynamic alterations among immune cells are critical in shaping the tumor microenvironment and ultimately map onto heterogeneous clinical outcomes. Currently, the underlying clinical significance of immune cell evolutions remains largely unexplored in hepatocellular carcinoma (HCC). Methods A total of 3,817 immune cells and 1,750 HCC patients of 15 independent public datasets were retrieved. The Seurat and Monocle algorithms were used to depict T cell evolution, and nonnegative matrix factorization (NMF) was further applied to identify the molecular classification. Subsequently, the prognosis, biological characteristics, genomic variations, and immune landscape among distinct clusters were decoded. The clinical efficacy of multiple treatment approaches was further investigated. Results According to trajectory gene expression, three heterogeneous clusters with different clinical outcomes were identified. C2, with a more advanced pathological stage, presented the most dismal prognosis relative to C1 and C3. Eight independent external cohorts validated the robustness and reproducibility of the three clusters. Further explorations elucidated C1 to be characterized as lipid metabolic HCC, and C2 was referred to as cell-proliferative HCC, whereas C3 was defined as immune inflammatory HCC. Moreover, C2 also displayed the most conspicuous genomic instability, and C3 was deemed as “immune-hot”, having abundant immune cells and an elevated expression of immune checkpoints. The assessments of therapeutic intervention suggested that patients in C1 were suitable for transcatheter arterial chemoembolization treatment, and patients in C2 were sensitive to tyrosine kinase inhibitors, while patients in C3 were more responsive to immunotherapy. We also identified numerous underlying therapeutic agents, which might be conducive to clinical transformation in the future. Conclusions Our study developed three clusters with distinct characteristics based on immune cell evolutions. For specifically stratified patients, we proposed individualized treatment strategies to improve the clinical outcomes and facilitate the clinical management.
Collapse
Affiliation(s)
- Long Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Diagnosis and Treatment League for Hepatopathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering and Research Center for Diagnosis and Treatment of Hepatobiliary and Pancreatic Surgical Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Gao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Diagnosis and Treatment League for Hepatopathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering and Research Center for Diagnosis and Treatment of Hepatobiliary and Pancreatic Surgical Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xudong Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Diagnosis and Treatment League for Hepatopathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering and Research Center for Diagnosis and Treatment of Hepatobiliary and Pancreatic Surgical Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunguang Guo
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bowen Hu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Diagnosis and Treatment League for Hepatopathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering and Research Center for Diagnosis and Treatment of Hepatobiliary and Pancreatic Surgical Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhihui Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Diagnosis and Treatment League for Hepatopathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering and Research Center for Diagnosis and Treatment of Hepatobiliary and Pancreatic Surgical Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiakai Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Diagnosis and Treatment League for Hepatopathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering and Research Center for Diagnosis and Treatment of Hepatobiliary and Pancreatic Surgical Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jihua Shi
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Diagnosis and Treatment League for Hepatopathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering and Research Center for Diagnosis and Treatment of Hepatobiliary and Pancreatic Surgical Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Diagnosis and Treatment League for Hepatopathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering and Research Center for Diagnosis and Treatment of Hepatobiliary and Pancreatic Surgical Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Diagnosis and Treatment League for Hepatopathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering and Research Center for Diagnosis and Treatment of Hepatobiliary and Pancreatic Surgical Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Shuijun Zhang,
| |
Collapse
|
20
|
Lan F, Chen X, Xiong Z, Cao Z, Lu L, Zhong Y, Zhan X, Yang Y, Shao Y, Li M, Han Z, Zhu X. Comprehensive transcriptomic and co-expression analysis of ABL1 gene and molecularly targeted drugs in hepatocellular carcinoma based on multi-database mining. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:146. [PMID: 35834027 DOI: 10.1007/s12032-022-01730-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/05/2022] [Indexed: 11/28/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death worldwide. Consequently, it is essential to identify biomarkers for treatment response and the prognosis prediction. We investigated whether ABL1 can function as a biomarker or a drug target for HCC. We assessed the ABL1 expression, genetic alterations and patients' survival from LinkedOmics, GEO, TCGA and Human Protein Atlas. We analyzed PPI, GO and KEGG pathways. GSEA was analyzed for functional comparison. The current drugs targeting ABL1 were statistically analyzed using DRUGSURV and DGIdb database. We found ABL1 is overexpressed in HCC and its higher expression reduces survival probability. Genetic changes of ABL1 are not frequent. We screened out 25 differentially expressed genes correlated with ABL1. The top functions of ABL1 are biological regulation, metabolic process, protein-containing, and protein binding. KEGG pathways showed that ABL1 and correlated with ABL1 significantly genes markedly enriched in the ErbB signaling pathway, and pathways in cancer. We counted the existing drugs targeting ABL1, which indicates that inhibiting ABL1 expression may improve the survival probability of HCC. In conclusion, ABL1 plays a crucial role in the development and progression of this cancerization and is a potential drug target.
Collapse
Affiliation(s)
- Feifei Lan
- Medical Genetics Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Xinqia Chen
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Zhuolong Xiong
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Zitong Cao
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Liangzong Lu
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Yueyuan Zhong
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Xuliang Zhan
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Yue Yang
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Yingqi Shao
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Minhua Li
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Zenglei Han
- Department of Pathology, Qingdao Municipal Hospital, Qingdao, China.
| | - Xiao Zhu
- Zhu's Team, Guangdong Medical University, Zhanjiang, China. .,School of Laboratory Medicine and Biomedical Engineering, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
21
|
Hou W, Bridgeman B, Malnassy G, Ding X, Cotler S, Dhanarajan A, Qiu W. Integrin subunit beta 8 contributes to lenvatinib resistance in HCC. Hepatol Commun 2022; 6:1786-1802. [PMID: 35238496 PMCID: PMC9234648 DOI: 10.1002/hep4.1928] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/08/2022] [Accepted: 02/13/2022] [Indexed: 11/11/2022] Open
Abstract
Lenvatinib is a multikinase inhibitor approved as a first-line therapy for advanced hepatocellular carcinoma (HCC). However, the development of drug resistance is common, and the underlying mechanisms governing this resistance are largely unknown. In this study, we established two lenvatinib-resistant (LR) HCC cell lines and identified integrin subunit beta 8 (ITGB8) as a critical contributor to lenvatinib resistance in HCC. The elevated expression of ITGB8 was observed in LR HCC cells. Furthermore, silencing of ITGB8 reversed lenvatinib resistance in vitro and in vivo, whereas ectopic expression of ITGB8 in lenvatinib-sensitive parental HCC cells exhibited increased resistance to lenvatinib. Mechanistically, ITGB8 regulated lenvatinib resistance through an HSP90-mediated stabilization of AKT and enhanced AKT signaling. In support of this model, either an AKT inhibitor MK-2206 or an HSP90 inhibitor 17-AAG resensitized LR HCC cells to lenvatinib treatment. Conclusion: Collectively, our results establish a crucial role of ITGB8 in lenvatinib resistance, and suggest that targeting the ITGB8/HSP90/AKT axis is a promising therapeutic strategy in patients with HCC exhibiting lenvatinib resistance.
Collapse
Affiliation(s)
- Wei Hou
- Department of SurgeryLoyola University Chicago Stritch School of MedicineMaywoodIllinoisUSA
- Department of Cancer BiologyLoyola University Chicago Stritch School of MedicineMaywoodIllinoisUSA
| | - Bryan Bridgeman
- Department of SurgeryLoyola University Chicago Stritch School of MedicineMaywoodIllinoisUSA
- Department of Cancer BiologyLoyola University Chicago Stritch School of MedicineMaywoodIllinoisUSA
| | - Greg Malnassy
- Department of SurgeryLoyola University Chicago Stritch School of MedicineMaywoodIllinoisUSA
- Department of Cancer BiologyLoyola University Chicago Stritch School of MedicineMaywoodIllinoisUSA
| | - Xianzhong Ding
- Department of PathologyLoyola University Chicago Stritch School of MedicineMaywoodIllinoisUSA
| | - Scott J. Cotler
- Department of MedicineLoyola University Chicago Stritch School of MedicineMaywoodIllinoisUSA
| | - Asha Dhanarajan
- Department of MedicineLoyola University Chicago Stritch School of MedicineMaywoodIllinoisUSA
| | - Wei Qiu
- Department of SurgeryLoyola University Chicago Stritch School of MedicineMaywoodIllinoisUSA
- Department of Cancer BiologyLoyola University Chicago Stritch School of MedicineMaywoodIllinoisUSA
| |
Collapse
|
22
|
Zou X, Tang XY, Qu ZY, Sun ZW, Ji CF, Li YJ, Guo SD. Targeting the PDGF/PDGFR signaling pathway for cancer therapy: A review. Int J Biol Macromol 2022; 202:539-557. [PMID: 35074329 DOI: 10.1016/j.ijbiomac.2022.01.113] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023]
Abstract
Platelet-derived growth factors (PDGFs) and PDGF receptors (PDGFRs) are expressed in a variety of tumors. Activation of the PDGF/PDGFR signaling pathway is associated with cancer proliferation, metastasis, invasion, and angiogenesis through modulating multiple downstream pathways, including phosphatidylinositol 3 kinase/protein kinase B pathway and mitogen-activated protein kinase/extracellular signal-regulated kinase pathway. Therefore, targeting PDGF/PDGFR signaling pathway has been demonstrated to be an effective strategy for cancer therapy, and accordingly, some great progress has been made in this field in the past few decades. This review will focus on the PDGF isoforms and their binding with the related PDGFRs, the PDGF/PDGFR signaling and regulation, and especially present strategies and inhibitors developed for cancer therapy, and the related clinical benefits and side effects.
Collapse
Affiliation(s)
- Xiang Zou
- Engineering Research Center of Natural Antineoplastic Drugs, Ministry of Education, Harbin University of Commerce, 150076, China
| | - Xi-Yu Tang
- Engineering Research Center of Natural Antineoplastic Drugs, Ministry of Education, Harbin University of Commerce, 150076, China
| | - Zhong-Yuan Qu
- School of Pharmacy, Harbin University of Commerce, Harbin 150076, China.
| | - Zhi-Wei Sun
- School of Pharmacy, Harbin University of Commerce, Harbin 150076, China
| | - Chen-Feng Ji
- Engineering Research Center of Natural Antineoplastic Drugs, Ministry of Education, Harbin University of Commerce, 150076, China
| | - Yan-Jie Li
- Institute of lipid metabolism and Atherosclerosis, School of Pharmacy, Weifang Medical University, Weifang 261053, China.
| | - Shou-Dong Guo
- Engineering Research Center of Natural Antineoplastic Drugs, Ministry of Education, Harbin University of Commerce, 150076, China; School of Pharmacy, Harbin University of Commerce, Harbin 150076, China; Institute of lipid metabolism and Atherosclerosis, School of Pharmacy, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
23
|
Xu H, Qin J, Qin L, Guo C, Yang B. Bioinformatics and In Silico Findings Uncover Bio-Targets of Calycosin Against Heart Failure and Diabetes Mellitus. Front Endocrinol (Lausanne) 2022; 13:790619. [PMID: 35898453 PMCID: PMC9309256 DOI: 10.3389/fendo.2022.790619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 05/23/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Heart failure (HF) and diabetes mellitus (DM) are life-threatening diseases. However, existing clinical drugs to treat HF complicated with DM are relatively limited. In this study, we performed a viable bioinformatics strategy combining network pharmacology and molecular docking to identify potential anti-HF and -DM targets and therapeutic mechanisms of calycosin, a functional phytoestrogen. METHODS Web-based databases were used to collect candidate genes/targets of calycosin and HF/DM and then identify the hub bio-targets of calycosin against HF/DM. Using the online-available database, all functional processes and signaling pathways of calycosin against HF/DM were screened and identified before further visualization. RESULTS All potential bio-targets of calycosin and HF/DM were collected, and 20 hub targets of calycosin against HF/DM were identified. Interestingly, molecular docking findings indicated that mitogen-activated protein kinase-1 (MAPK1), β-arrestin 1 (ARRB1), and homologue-1 (ABL1) may be potent pharmacological targets of calycosin against HF/DM. In addition, all primary molecular functions of calycosin against HF/DM were identified, including regulating protein binding, ubiquitination, and the metabolic process. Furthermore, the top molecular pathways of calycosin against HF/DM were revealed, including cardiomyocyte and chemokine signaling pathways. CONCLUSION Our bioinformatics analysis uncovered the network targets and therapeutic mechanisms of calycosin against HF/DM. For the first time, the current in silico findings revealed that the identified hub targets may be used to screen and treat HF/DM.
Collapse
Affiliation(s)
- Hongyuan Xu
- Cardiology Department, Guigang City People’s Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, China
| | - Jingru Qin
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Lixiu Qin
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Chao Guo
- Department of Pharmacy, Guigang City People’s Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, China
- *Correspondence: Chao Guo, ; Bin Yang,
| | - Bin Yang
- College of Pharmacy, Guangxi Medical University, Nanning, China
- *Correspondence: Chao Guo, ; Bin Yang,
| |
Collapse
|
24
|
Abstract
Hepatocellular carcinoma (HCC) is a very deadly disease. HCC initiation and progression involve multiple genetic events, including the activation of proto-oncogenes and disruption of the function of specific tumor suppressor genes. Activation of oncogenes stimulates cell growth and survival, while loss-of-function mutations of tumor suppressor genes result in unrestrained cell growth. In this review, we summarize the new findings that identified novel proto-oncogenes and tumor suppressors in HCC over the past five years. These findings may inspire the development of novel therapeutic strategies to improve the outcome of HCC patients.
Collapse
|
25
|
ABL1 Is a Prognostic Marker and Associated with Immune Infiltration in Hepatocellular Carcinoma. JOURNAL OF ONCOLOGY 2021; 2021:1379706. [PMID: 34484330 PMCID: PMC8413061 DOI: 10.1155/2021/1379706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/02/2021] [Accepted: 08/06/2021] [Indexed: 12/23/2022]
Abstract
Background The role of ABL1 in hepatocellular carcinoma (HCC) is still unclear. Therefore, this study aims to explore the potential role of ABL1 in the progression of HCC using bioinformatics methods. Methods We analyzed the expression, prognostic potential, and immune cell effect of ABL1 in HCC by using a variety of datasets. Results ABL1 is highly expressed in HCC and associated with unfavorable overall survival (OS) and disease-free survival (DFS). Functional network analysis revealed that ABL1 plays an important role in mitochondrial activity, ATP metabolism, protein translation and metabolism, various neurological diseases, nonalcoholic fatty liver disease, and notch signaling pathway. In addition, we found that ABL1 expression was closely correlated with B cells, CD8 + T cells, CD4 + T cells, macrophages, neutrophils, and dendritic cells. Furthermore, ABL1 expression was positively associated with the expression levels of immune checkpoint genes, such as PD-1L, TIM3, TIGIT, and CTLA4. Conclusion ABL1 is associated with immune infiltration and prognosis of HCC.
Collapse
|
26
|
Wang P, Sun Y, Jin W, Cheng J, Peng H, Xu Y, Zhou K, Chen L, Huang K, Wu S, Hu B, Zhang Z, Guo W, Cao Y, Zhou J, Fan J, Yang X. Circulating tumor cell detection and single-cell analysis using an integrated workflow based on ChimeraX ® -i120 Platform: A prospective study. Mol Oncol 2021; 15:2345-2362. [PMID: 33301640 PMCID: PMC8410565 DOI: 10.1002/1878-0261.12876] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/16/2020] [Accepted: 12/03/2020] [Indexed: 12/23/2022] Open
Abstract
Circulating tumor cell (CTC) analysis holds great potential to be a noninvasive solution for clinical cancer management. A complete workflow that combined CTC detection and single-cell molecular analysis is required. We developed the ChimeraX® -i120 platform to facilitate negative enrichment, immunofluorescent labeling, and machine learning-based identification of CTCs. Analytical performances were evaluated, and a total of 477 participants were enrolled to validate the clinical feasibility of ChimeraX® -i120 CTC detection. We analyzed copy number alteration profiles of isolated single cells. The ChimeraX® -i120 platform had high sensitivity, accuracy, and reproducibility for CTC detection. In clinical samples, an average value of > 60% CTC-positive rate was found for five cancer types (i.e., liver, biliary duct, breast, colorectal, and lung), while CTCs were rarely identified in blood from healthy donors. In hepatocellular carcinoma patients treated with curative resection, CTC status was significantly associated with tumor characteristics, prognosis, and treatment response (all P < 0.05). Single-cell sequencing analysis revealed that heterogeneous genomic alteration patterns resided in different cells, patients, and cancers. Our results suggest that the use of this ChimeraX® -i120 platform and the integrated workflow has validity as a tool for CTC detection and downstream genomic profiling in the clinical setting.
Collapse
Affiliation(s)
- Peng‐Xiang Wang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Yun‐Fan Sun
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | | | - Jian‐Wen Cheng
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | | | - Yang Xu
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Kai‐Qian Zhou
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | | | | | - Sui‐Yi Wu
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Bo Hu
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Ze‐Fan Zhang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Wei Guo
- Department of Laboratory MedicineZhongshan HospitalFudan UniversityShanghaiChina
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Cancer InvasionCancer Research InstituteCentral South UniversityMinistry of EducationChangshaChina
| | - Jian Zhou
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Jia Fan
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Xin‐Rong Yang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| |
Collapse
|
27
|
Sun Y, Tang X, Ye B, Ding K. DNA and RNA Sequencing Recapitulated Aberrant Tumor Metabolism in Liver Cancer Cell Lines. J Hepatocell Carcinoma 2021; 8:823-836. [PMID: 34350138 PMCID: PMC8327295 DOI: 10.2147/jhc.s318724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022] Open
Abstract
AIM Metabolic reprogramming has recently attracted extensive attention for understanding cancer development. We aimed to demonstrate a genomic and transcriptomic landscape of metabolic reprogramming underlying liver cancer cell lines. METHODS We investigated metabolic aberrant at both the transcriptome and genome levels using transcriptome and whole-exome sequencing data from 12 human liver cancer cell lines (hLCCLs) and one normal liver cell line. RESULTS Three subgroups of hLCCLs characterized from transcriptome sequencing data exhibit significantly different aberrations in various metabolic processes, including amino acid, lipid, energy, and carbohydrate metabolism. Furthermore, whole-exome sequencing revealed distinct mutational signatures among different subgroups of hLCCLs and identified a total of 19 known driver genes implicated in metabolism. CONCLUSION Our findings highlighted differential metabolic mechanisms in the development of liver cancer and provided a resource for further investigating its metabolic mechanisms.
Collapse
Affiliation(s)
- Yihong Sun
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, 410006, People’s Republic of China
| | - Xia Tang
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, 410006, People’s Republic of China
| | - Bo Ye
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, 410006, People’s Republic of China
| | - Keyue Ding
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, 410006, People’s Republic of China
- Medical Genetic Institute of Henan Province, Henan Provincial People’s Hospital, Henan Key Laboratory of Genetic Diseases and Functional Genomics, National Health Commission Key Laboratory of Birth Defect Prevention, Henan Provincial People’s Hospital of Henan University, People’s Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450003, People's Republic of China
| |
Collapse
|
28
|
Luttman JH, Colemon A, Mayro B, Pendergast AM. Role of the ABL tyrosine kinases in the epithelial-mesenchymal transition and the metastatic cascade. Cell Commun Signal 2021; 19:59. [PMID: 34022881 PMCID: PMC8140471 DOI: 10.1186/s12964-021-00739-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
The ABL kinases, ABL1 and ABL2, promote tumor progression and metastasis in various solid tumors. Recent reports have shown that ABL kinases have increased expression and/or activity in solid tumors and that ABL inactivation impairs metastasis. The therapeutic effects of ABL inactivation are due in part to ABL-dependent regulation of diverse cellular processes related to the epithelial to mesenchymal transition and subsequent steps in the metastatic cascade. ABL kinases target multiple signaling pathways required for promoting one or more steps in the metastatic cascade. These findings highlight the potential utility of specific ABL kinase inhibitors as a novel treatment paradigm for patients with advanced metastatic disease. Video abstract.
Collapse
Affiliation(s)
- Jillian Hattaway Luttman
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| | - Ashley Colemon
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| | - Benjamin Mayro
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| | - Ann Marie Pendergast
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| |
Collapse
|