1
|
Ding M, Mao S, Wu H, Fang S, Zhen N, Chen T, Zhu J, Tang X, Wang X, Sun F, Zhu G, Pan Q, Ma J. Malignant Hepatoblast-Like Cells Sustain Stemness via IGF2-Dependent Cholesterol Accumulation in Hepatoblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407671. [PMID: 40271711 PMCID: PMC12120738 DOI: 10.1002/advs.202407671] [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] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 02/08/2025] [Indexed: 04/25/2025]
Abstract
Hepatoblastoma, the most aggressive childhood liver tumor, poses significant challenges due to limited knowledge of its pathogenesis, particularly in poorly differentiated advanced tumors where the prognosis is dismal. Single-cell sequencing provides an in-depth exploration at the single-cell level and offers a deep understanding of tumor heterogeneity. Herein, single-cell transcriptomics analysis is used to identify a unique malignant-hepatoblast (HB)-like cell subpopulation as the possible origin of poorly differentiated hepatoblastoma. These cells are associated with an unfavorable clinical prognosis in hepatoblastoma patients. The malignant-HB-like cell subpopulation generated insulin-like growth factor 2 (IGF2) to sustain stem-like features by promoting abnormal cholesterol accumulation via SREBF2. IGF2 also stimulated fibroblast 2 to secrete collagen 1, intensifying tumor malignancy via the collagen 1/integrin α1 signaling pathway. This suggests that targeting malignant HB-like cells by inhibiting IGF2-induced pathways can lead to promising treatments for hepatoblastoma. Additionally, serum IGF2 levels may serve as a diagnostic biomarker for advanced hepatoblastoma. In summary, these findings provide valuable insight into the genesis and malignancy of hepatoblastoma and a foundation for more effective diagnostic tools and therapeutic strategies for this challenging disease.
Collapse
Affiliation(s)
- Miao Ding
- Department of Clinical LaboratoryShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200120P. R. China
| | - Siwei Mao
- Department of Clinical LaboratoryShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200120P. R. China
| | - Han Wu
- Department of Clinical LaboratoryShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200120P. R. China
| | - Sijia Fang
- Department of Clinical LaboratoryShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200120P. R. China
| | - Ni Zhen
- Department of Clinical LaboratoryShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200120P. R. China
| | - Tianshu Chen
- Department of Clinical LaboratoryShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200120P. R. China
| | - Jiabei Zhu
- Department of Clinical LaboratoryShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200120P. R. China
| | - Xiaochen Tang
- Department of Clinical LaboratoryShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200120P. R. China
| | - Xiaoyang Wang
- Department of Clinical LaboratoryShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200120P. R. China
| | - Fenyong Sun
- Department Laboratory MedicineShanghai Tenth People's Hospital of Tongji UniversityShanghai200072P. R. China
| | - Guoqing Zhu
- Department of Clinical LaboratoryShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200120P. R. China
| | - Qiuhui Pan
- Department of Clinical LaboratoryShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200120P. R. China
- Faculty of Medical Laboratory ScienceCollege of Health Science and TechnologyShanghai Jiao Tong University School of MedicineShanghai200000P. R. China
- Shanghai Key Laboratory of Clinical Molecular Diagnostics for PediatricsShanghai200120P. R. China
- Sanya Women and Children's Hospital Managed by Shanghai Children's Medical CenterSanya572029P. R. China
| | - Ji Ma
- Department of Clinical LaboratoryShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200120P. R. China
- Shanghai Key Laboratory of Clinical Molecular Diagnostics for PediatricsShanghai200127P. R. China
| |
Collapse
|
2
|
Chen R, Chen T, Li X, Yu J, Lin M, Wen S, Zhang M, Chen J, Yi B, Zhong H, Li Z. SREBP2 as a central player in cancer progression: potential for targeted therapeutics. Front Pharmacol 2025; 16:1535691. [PMID: 40308757 PMCID: PMC12041066 DOI: 10.3389/fphar.2025.1535691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Recent studies have identified the reprogramming of lipid metabolism as a critical hallmark of malignancy. Enhanced cholesterol uptake and increased cholesterol biosynthesis significantly contribute to the rapid growth of tumors, with cholesterol also playing essential roles in cellular signaling pathways. Targeting cholesterol metabolism has emerged as a promising therapeutic strategy in oncology. The sterol regulatory element-binding protein-2 (SREBP2) serves as a primary transcriptional regulator of genes involved in cholesterol biosynthesis and is crucial for maintaining cholesterol homeostasis. Numerous studies have reported the upregulation of SREBP2 across various cancers, facilitating tumor progression. This review aims to provide a comprehensive overview of the structure, biological functions, and regulatory mechanisms of SREBP2. Furthermore, we summarize that SREBP2 plays a crucial role in various cancers and tumor microenvironment primarily by regulating cholesterol, as well as through several non-cholesterol pathways. We also particularly emphasize therapeutic agents targeting SREBP2 that are currently under investigation. This review seeks to enhance our understanding of SREBP2's involvement in cancer and provide theoretical references for cancer therapies that target SREBP2.
Collapse
Affiliation(s)
- Ruiqi Chen
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Tianyu Chen
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiang Li
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Junfeng Yu
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Min Lin
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Siqi Wen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Man Zhang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jinchi Chen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Bei Yi
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Huage Zhong
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, China
| | - Zhao Li
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
3
|
Deng H, Wang X, Jiang ZA, Xu J, Zhang Y, Zhou Y, Gong J, Lu XY, Hou YF, Zhang H. Clinical potential and experimental validation of prognostic genes in hepatocellular carcinoma revealed by risk modeling utilizing single cell and transcriptome constructs. Front Immunol 2025; 16:1541252. [PMID: 40255404 PMCID: PMC12006083 DOI: 10.3389/fimmu.2025.1541252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/03/2025] [Indexed: 04/22/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the leading cause of tumor-related mortality worldwide. There is an urgent need for predictive biomarkers to guide treatment decisions. This study aimed to identify robust prognostic genes for HCC and to establish a theoretical foundation for clinical interventions. Methods The HCC datasets were obtained from public databases and then differential expression analysis were used to obtain significant gene expression profiles. Subsequently, univariate Cox regression analysis and PH assumption test were performed, and a risk model was developed using an optimal algorithm from 101 combinations on the TCGA-LIHC dataset to pinpoint prognostic genes. Immune infiltration and drug sensitivity analyses were conducted to assess the impact of these genes and to explore potential chemotherapeutic agents for HCC. Additionally, single-cell analysis was employed to identify key cellular players and their interactions within the tumor microenvironment. Finally, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was utilized to validate the roles of these prognostic genes in HCC. Results A total of eight prognostic genes were identified (MCM10, CEP55, KIF18A, ORC6, KIF23, CDC45, CDT1, and PLK4). The risk model, constructed based on these genes, was effective in predicting survival outcomes for HCC patients. CEP55 exhibited the strongest positive correlation with activated CD4 T cells. The top 10 drugs showed increased sensitivity in the low-risk group. B cells were identified as key cellular components with the highest interaction numbers and strengths with macrophages in both HCC and control groups. Prognostic genes were more highly expressed in the initial state of B cell differentiation. RT-qPCR confirmed significant upregulation of MCM10, KIF18A, CDC45, and PLK4 in HCC tissues (p< 0.05). Conclusion This study successfully identified eight prognostic genes (MCM10, CEP55, KIF18A, ORC6, KIF23, CDC45, CDT1, and PLK4), which provided new directions for exploring the potential pathogenesis and clinical treatment research of HCC.
Collapse
Affiliation(s)
- Hang Deng
- Medical College, University of Electronic Science and Technology of China, Chengdu, China
| | - Xu Wang
- Department of Hepatobiliary Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Zi-Ang Jiang
- Medical College, North Sichuan Medical College, Nanchong, China
| | - Jian Xu
- Department of Hepatobiliary Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Zhang
- Department of Hepatobiliary Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yao Zhou
- Department of Hepatobiliary Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jun Gong
- Department of Hepatobiliary Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiang-Yu Lu
- Department of Hepatobiliary Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi-Fu Hou
- Department of Organ Translation Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hao Zhang
- Department of Hepatobiliary Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
4
|
Hu H, Zhang Y, Yu Y, Liu D, Dong Z, Chen G. Phosphoproteomic analysis of X-ray-irradiated planarians provides novel insights into the DNA damage response. Int J Biol Macromol 2025; 299:140129. [PMID: 39842578 DOI: 10.1016/j.ijbiomac.2025.140129] [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: 11/06/2024] [Revised: 01/10/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
Phosphorylation plays a crucial role in the cellular response to radiation and cancer therapies, yet phosphoproteomics studies in planarians remain underexplored despite the organism's remarkable regenerative capacities. This study utilized advanced ion mobility mass spectrometry for 4D-label-free quantitative proteomics to identify phosphorylation sites associated with irradiation in planarians. A total of 33,284 phosphorylation sites from 15,505 phosphorylated peptides and 4710 unique phosphoproteins were identified. In the sub-lethal dose irradiation group, 1695 phosphoproteins with 3483 phosphorylation sites exhibited significant changes, while exposure to lethal doses of radiation led to significant changes in 2308 phosphoproteins with 6112 phosphorylation sites, including many kinases, transcription factors, and cytoskeletal proteins. Functional enrichment analysis revealed that the altered phosphoproteins were primarily involved in transcription, RNA biosynthesis, mRNA processing regulation, and spliceosomal complex assembly. Functional validation of five differentially phosphorylated proteins revealed that their depletion impaired stem cell regeneration after irradiation by disrupting DNA repair, suggesting that these proteins are critical to planarian biology and their radiation response. By identifying the phosphorylation state and specific sites of planarian proteins, our study lays the foundation for further research on protein phosphorylation in the radiation-induced DNA damage response. In addition, our findings provide preliminary insights into the role of calnexin, a protein involved in interacting with newly synthesized N-linked glycoproteins, in planarians.
Collapse
Affiliation(s)
- Huanhuan Hu
- College of Life Science, Henan Normal University, Xinxiang 453007, Henan Province, PR China; Key Laboratory of Fertility Preservation, School of Life Sciences and Technologies, Sanquan College of Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Yibing Zhang
- College of Life Science, Henan Normal University, Xinxiang 453007, Henan Province, PR China
| | - Yanan Yu
- College of Life Science, Henan Normal University, Xinxiang 453007, Henan Province, PR China
| | - Dezeng Liu
- College of Life Science, Henan Normal University, Xinxiang 453007, Henan Province, PR China
| | - Zimei Dong
- College of Life Science, Henan Normal University, Xinxiang 453007, Henan Province, PR China.
| | - Guangwen Chen
- College of Life Science, Henan Normal University, Xinxiang 453007, Henan Province, PR China.
| |
Collapse
|
5
|
Sugimoto A, Saito Y, Wang G, Sun Q, Yin C, Lee KH, Geng Y, Rajbhandari P, Hernandez C, Steffani M, Qie J, Savage T, Goyal DM, Ray KC, Neelakantan TV, Yin D, Melms J, Lehrich BM, Yasaka TM, Liu S, Oertel M, Lan T, Guillot A, Peiseler M, Filliol A, Kanzaki H, Fujiwara N, Ravi S, Izar B, Brosch M, Hampe J, Remotti H, Argemi J, Sun Z, Kendall TJ, Hoshida Y, Tacke F, Fallowfield JA, Blockley-Powell SK, Haeusler RA, Steinman JB, Pajvani UB, Monga SP, Bataller R, Masoodi M, Arpaia N, Lee YA, Stockwell BR, Augustin HG, Schwabe RF. Hepatic stellate cells control liver zonation, size and functions via R-spondin 3. Nature 2025; 640:752-761. [PMID: 40074890 PMCID: PMC12003176 DOI: 10.1038/s41586-025-08677-w] [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: 03/04/2024] [Accepted: 01/21/2025] [Indexed: 03/14/2025]
Abstract
Hepatic stellate cells (HSCs) have a central pathogenetic role in the development of liver fibrosis. However, their fibrosis-independent and homeostatic functions remain poorly understood1-5. Here we demonstrate that genetic depletion of HSCs changes WNT activity and zonation of hepatocytes, leading to marked alterations in liver regeneration, cytochrome P450 metabolism and injury. We identify R-spondin 3 (RSPO3), an HSC-enriched modulator of WNT signalling, as responsible for these hepatocyte-regulatory effects of HSCs. HSC-selective deletion of Rspo3 phenocopies the effects of HSC depletion on hepatocyte gene expression, zonation, liver size, regeneration and cytochrome P450-mediated detoxification, and exacerbates alcohol-associated and metabolic dysfunction-associated steatotic liver disease. RSPO3 expression decreases with HSC activation and is inversely associated with outcomes in patients with alcohol-associated and metabolic dysfunction-associated steatotic liver disease. These protective and hepatocyte-regulating functions of HSCs via RSPO3 resemble the R-spondin-expressing stromal niche in other organs and should be integrated into current therapeutic concepts.
Collapse
Affiliation(s)
- Atsushi Sugimoto
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Yoshinobu Saito
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Guanxiong Wang
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Qiuyan Sun
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Chuan Yin
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Ki Hong Lee
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yana Geng
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Presha Rajbhandari
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA
| | - Celine Hernandez
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Marcella Steffani
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Jingran Qie
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Thomas Savage
- Department of Microbiology & Immunology, Columbia University, New York, NY, USA
| | - Dhruv M Goyal
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Kevin C Ray
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Taruna V Neelakantan
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA
| | - Deqi Yin
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Johannes Melms
- Department of Medicine, Columbia University, New York, NY, USA
| | - Brandon M Lehrich
- Department of Pharmacology and Chemical Biology, Pittsburgh Liver Research Center, and Organ Pathobiology and Therapeutics Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tyler M Yasaka
- Department of Pharmacology and Chemical Biology, Pittsburgh Liver Research Center, and Organ Pathobiology and Therapeutics Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Silvia Liu
- Department of Pharmacology and Chemical Biology, Pittsburgh Liver Research Center, and Organ Pathobiology and Therapeutics Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael Oertel
- Department of Pharmacology and Chemical Biology, Pittsburgh Liver Research Center, and Organ Pathobiology and Therapeutics Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tian Lan
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Adrien Guillot
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Moritz Peiseler
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Aveline Filliol
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Hiroaki Kanzaki
- Liver Tumour Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Naoto Fujiwara
- Liver Tumour Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Samhita Ravi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Benjamin Izar
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Mario Brosch
- Department of Internal Medicine I, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jochen Hampe
- Department of Internal Medicine I, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Helen Remotti
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Josepmaria Argemi
- Liver Unit and RNA Biology and Therapies Program, Cima Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, Madrid, Spain
| | - Zhaoli Sun
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Timothy J Kendall
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Yujin Hoshida
- Liver Tumour Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Storm K Blockley-Powell
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Rebecca A Haeusler
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | | | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
- Institute of Human Nutrition, New York, NY, USA
| | - Satdarshan P Monga
- Department of Pharmacology and Chemical Biology, Pittsburgh Liver Research Center, and Organ Pathobiology and Therapeutics Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ramon Bataller
- Liver Unit,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain
| | - Mojgan Masoodi
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Nicholas Arpaia
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Youngmin A Lee
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brent R Stockwell
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany.
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| | - Robert F Schwabe
- Department of Medicine, Columbia University, New York, NY, USA.
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA.
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Institute of Human Nutrition, New York, NY, USA.
- Burch-Lodge Center for Human Longevity, Columbia University, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, New York, NY, USA.
| |
Collapse
|
6
|
Wu Y, Song W, Su M, He J, Hu R, Zhao Y. The Role of Cholesterol Metabolism and Its Regulation in Tumor Development. Cancer Med 2025; 14:e70783. [PMID: 40145543 PMCID: PMC11948085 DOI: 10.1002/cam4.70783] [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: 10/28/2024] [Revised: 02/27/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Within the tumor microenvironment, tumor cells undergo metabolic reprogramming of cholesterol due to intrinsic cellular alterations and changes in the extracellular milieu. Furthermore, cholesterol reprogramming within this microenvironment influences the immune landscape of tumors, facilitating immune evasion and consequently promoting tumorigenesis. These biological changes involve modifications in numerous enzymes associated with cholesterol uptake and synthesis, including NPC1L1, SREBP, HMGCR, SQLE, and PCSK9. REVIEW This review systematically summarizes the role of cholesterol metabolism and its associated enzymes in cancer progression, examines the mechanisms through which dysregulation of cholesterol metabolism affects immune cells within the tumor microenvironment, and discusses recent advancements in cancer therapies that target cholesterol metabolism. CONCLUSION Targeting cholesterol metabolism-related enzymes can inhibit tumor growth, reshape immune landscapes, and rejuvenate antitumor immunity, offering potential therapeutic avenues in cancer treatment.
Collapse
Affiliation(s)
- Yongmei Wu
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Wenqian Song
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Min Su
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory of Regenerative Medicine in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Jing He
- Characteristic Key Laboratory of Translational Medicine Research of Cardiovascular and Cerebrovascular Diseases in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Rong Hu
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Characteristic Key Laboratory of Translational Medicine Research of Cardiovascular and Cerebrovascular Diseases in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Youbo Zhao
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory of Regenerative Medicine in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| |
Collapse
|
7
|
Ni Y, Liu B, Zhang W, Pang Y, Tian Y, Lv Q, Shi S, Zheng Y, Fan H. Evaluation of PDZD11 in hepatocellular carcinoma: prognostic value and diagnostic potential in combination with AFP. Front Oncol 2025; 15:1533865. [PMID: 40201341 PMCID: PMC11975663 DOI: 10.3389/fonc.2025.1533865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/06/2025] [Indexed: 04/10/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most prevalent liver cancer, with a 5-year survival rate below 20% and an average survival time of 3-6 months. Identifying new biomarkers is crucial for early diagnosis and prognosis. The function of PDZ domain protein 11 (PDZD11) in HCC remains unclear. Methods In this study, PDZD11 was investigated as a potential biomarker for HCC using bioinformatic analysis of the TCGA and ICGC datasets. Furthermore, we assessed the potential of serum PDZD11 as a clinical diagnostic marker by enrolling a cohort comprising 78 HCC patients and 62 healthy controls (HC) using the ELISA analysis and combining its expression with common tumor markers. Results Our research found significantly higher PDZD11 mRNA expression in HCC tissues compared to tumor-adjacent tissues (p < 0.001), which was associated with lower overall survival (OS) rates (p < 0.01). Multivariate evaluation methods established PDZD11 as a standalone predictor of prognosis. A nomogram incorporating PDZD11 expression and clinicopathological factors predicted OS rates for HCC patients over various years. Patients with HCC exhibited notably elevated serum PDZD11 levels compared to HC, with these levels rising further in advanced disease stages and deteriorating performance status (PS). ROC analysis showed high diagnostic accuracy when PDZD11 is combined with AFP (AUC = 0.958). Conclusion PDZD11 is more sensitive than AFP in assessing HCC prognosis. In conclusion, PDZD11 is a promising supplementary biomarker for HCC diagnosis and prognosis alongside AFP.
Collapse
Affiliation(s)
- Yiyun Ni
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Bin Liu
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Weizhen Zhang
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Yilin Pang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yaling Tian
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Qingsong Lv
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Shengwen Shi
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Yang Zheng
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Huihui Fan
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| |
Collapse
|
8
|
Zhong B, Du J, Liu F, Sun S. The Role of Yes-Associated Protein in Inflammatory Diseases and Cancer. MedComm (Beijing) 2025; 6:e70128. [PMID: 40066231 PMCID: PMC11892025 DOI: 10.1002/mco2.70128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 02/02/2025] [Accepted: 02/11/2025] [Indexed: 03/17/2025] Open
Abstract
Yes-associated protein (YAP) plays a central role in the Hippo pathway, primarily governing cell proliferation, differentiation, and apoptosis. Its significance extends to tumorigenesis and inflammatory conditions, impacting disease initiation and progression. Given the increasing relevance of YAP in inflammatory disorders and cancer, this study aims to elucidate its pathological regulatory functions in these contexts. Specifically, we aim to investigate the involvement and molecular mechanisms of YAP in various inflammatory diseases and cancers. We particularly focus on how YAP activation, whether through Hippo-dependent or independent pathways, triggers the release of inflammation and inflammatory mediators in respiratory, cardiovascular, and digestive inflammatory conditions. In cancer, YAP not only promotes tumor cell proliferation and differentiation but also modulates the tumor immune microenvironment, thereby fostering tumor metastasis and progression. Additionally, we provide an overview of current YAP-targeted therapies. By emphasizing YAP's role in inflammatory diseases and cancer, this study aims to enhance our understanding of the protein's pivotal involvement in disease processes, elucidate the intricate pathological mechanisms of related diseases, and contribute to future drug development strategies targeting YAP.
Collapse
Affiliation(s)
- Bing Zhong
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jintao Du
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Feng Liu
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Silu Sun
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesChinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and ManagementWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| |
Collapse
|
9
|
Castoldi M, Roy S, Angendohr C, Pellegrino R, Vucur M, Singer MT, Buettner V, Dille MA, Wolf SD, Heij LR, Ghallab A, Albrecht W, Hengstler JG, Flügen G, Knoefel WT, Bode JG, Zender L, Neumann UP, Heikenwälder M, Longerich T, Roderburg C, Luedde T. Regulation of KIF23 by miR-107 controls replicative tumor cell fitness in mouse and human hepatocellular carcinoma. J Hepatol 2025; 82:499-511. [PMID: 40235270 DOI: 10.1016/j.jhep.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 04/17/2025]
Abstract
BACKGROUND & AIMS In hepatocellular carcinoma (HCC), successful translation of experimental targets identified in mouse models to human patients has proven challenging. In this study, we used a comprehensive transcriptomic approach in mice to identify novel potential targets for therapeutic intervention in humans. METHODS We analyzed combined genome-wide miRNA and mRNA expression data in three pathogenically distinct mouse models of liver cancer. Effects of target genes on hepatoma cell fitness were evaluated by proliferation, survival and motility assays. TCGA and GEO databases, in combination with tissue microarrays, were used to validate the mouse targets and their impact on human HCC prognosis. Finally, the functional effects of the identified targets on tumorigenesis and tumor therapy were tested in hydrodynamic tail vein injection-based preclinical HCC models in vivo. RESULTS The expression of miR-107 was found to be significantly reduced in mouse models of liver tumors of various etiologies and in cohorts of humans with HCC. Overexpression of miR-107 or inhibition of its novel target kinesin family member 23 (Kif23) significantly reduced proliferation by interfering with cytokinesis, thereby controlling survival and motility of mouse and human hepatoma cells. In humans, KIF23 expression was found to be a prognostic marker in liver cancer, with high expression associated with poor prognosis. Hydrodynamic tail vein injection of vectors carrying either pre-miR-107 or anti-Kif23 shRNA inhibited the development of highly aggressive c-Myc-NRAS-induced liver cancers in mice. CONCLUSIONS Disruption of the miR-107/Kif23 axis inhibited hepatoma cell proliferation in vitro and prevented oncogene-induced liver cancer development in vivo, offering a novel potential avenue for the treatment of HCC in humans. IMPACT AND IMPLICATIONS Our study revealed the central role of the miR-107/KIF23 axis in controlling tumor cell fitness and hepatocellular carcinoma progression. The results demonstrate that the overexpression of miR-107 or silencing of its target, KIF23, markedly suppresses the proliferation, survival, and motility of human and mouse hepatoma cells. In this work, we demonstrate that the disruption of miR-107/Kif23 signaling effectively protects mice from an aggressive form of oncogene-induced liver cancer in vivo, implying that targeting miR-107/KIF23 might be a novel therapeutic approach for hepatocellular carcinoma in humans.
Collapse
Affiliation(s)
- Mirco Castoldi
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany.
| | - Sanchari Roy
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Carolin Angendohr
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Rossella Pellegrino
- Institute of Pathology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Mihael Vucur
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Michael T Singer
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Veronika Buettner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Matthias A Dille
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Stephanie D Wolf
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Lara R Heij
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany; Department of Surgery and Transplantation, University Hospital Essen, Essen, Germany; Department of Pathology, Erasmus Medical Center Rotterdam, The Netherlands
| | - Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, Dortmund, Germany; Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Wiebke Albrecht
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, Dortmund, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, Dortmund, Germany
| | - Georg Flügen
- Department of Surgery and Transplantation, University Hospital Essen, Essen, Germany; Department of Surgery, Heinrich-Heine-University and University Hospital Düsseldorf, Germany
| | - Wolfram T Knoefel
- Department of Surgery, Heinrich-Heine-University and University Hospital Düsseldorf, Germany
| | - Johannes G Bode
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Lars Zender
- Department of Internal Medicine VIII, University Hospital Tubingen, Tubingen, Germany
| | - Ulf P Neumann
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany; Department of Surgery and Transplantation, University Hospital Essen, Essen, Germany
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Longerich
- Institute of Pathology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Christoph Roderburg
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University Düsseldorf, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany.
| |
Collapse
|
10
|
Zhong X, Ott M, Sharma AD, Balakrishnan A. MicroRNA-107 - a small RNA with a big impact on cytokinesis in hepatocellular carcinoma. J Hepatol 2025; 82:414-416. [PMID: 39396645 DOI: 10.1016/j.jhep.2024.09.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024]
Affiliation(s)
- Xiaowei Zhong
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Research Group RNA Therapeutics & Liver Regeneration, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Michael Ott
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Amar Deep Sharma
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Research Group RNA Therapeutics & Liver Regeneration, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.
| | - Asha Balakrishnan
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
11
|
Cai D, Zhong G, Dai X, Zhao Z, Chen M, Hu J, Wu Z, Cheng L, Li S, Gong J. Targeting FDFT1 Reduces Cholesterol and Bile Acid Production and Delays Hepatocellular Carcinoma Progression Through the HNF4A/ALDOB/AKT1 Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411719. [PMID: 39899681 PMCID: PMC11948044 DOI: 10.1002/advs.202411719] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/17/2024] [Indexed: 02/05/2025]
Abstract
Targeting cholesterol metabolism is a novel direction for tumor therapy. Unfortunately, the current use of statins for hepatocellular carcinoma (HCC) is controversial. Herein, farnesyl-diphosphate farnesyltransferase 1 (FDFT1) is identified as a novel target for treating HCC and a potential alternative to statins. Twenty-three key genes in cholesterol biosynthesis are screened, and FDFT1 is identified via public databases (The Cancer Genome Atlas, International Cancer Genome Consortium and Gene Expression Omnibus). Clinical samples reveal that FDFT1 is highly expressed in HCC tissues, and this phenotype is strongly associated with a poor prognosis. Functionally, FDFT1 knockdown inhibits the proliferation and metastasis of HCC cells and suppresses hepatocarcinogenesis in vitro and in vivo, whereas FDFT1 overexpression promotes HCC cell proliferation and metastasis. Mechanistically, FDFT1 downregulation decreases cholesterol and bile acid levels and then increases hepatocyte nuclear factor 4 alpha (HNF4A) transcriptional activity. Experiments indicate that HNF4A combines with the promoter of aldolase B (ALDOB) and promotes the ALDOB transcription and that ALDOB combines with AKT serine/threonine kinase 1 (AKT1) and inhibits AKT1 phosphorylation. Moreover, FDFT1 knockdown combined with AKT inhibitor (AZD5363) treatment shows remarkable therapeutic potential. FDFT1 inhibition reduces cholesterol and bile acid levels to delay HCC progression through the HNF4A/ALDOB/AKT1 axis. Thus, targeting FDFT1 may be a novel potential strategy for treating HCC.
Collapse
Affiliation(s)
- Dong Cai
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Guo‐Chao Zhong
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Xin Dai
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Zhibo Zhao
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Menglin Chen
- Institute of Clinical PathologyKey Laboratory of Transplant Engineering and ImmunologyNHCWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Jiejun Hu
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Zhenru Wu
- Institute of Clinical PathologyKey Laboratory of Transplant Engineering and ImmunologyNHCWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Lve Cheng
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Shengwei Li
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Jianping Gong
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| |
Collapse
|
12
|
Sererols-Viñas L, Garcia-Vicién G, Ruiz-Blázquez P, Lee TF, Lee YA, Gonzalez-Sanchez E, Vaquero J, Moles A, Filliol A, Affò S. Hepatic Stellate Cells Functional Heterogeneity in Liver Cancer. Semin Liver Dis 2025; 45:33-51. [PMID: 40043738 DOI: 10.1055/a-2551-0724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Hepatic stellate cells (HSCs) are the liver's pericytes, and play key roles in liver homeostasis, regeneration, fibrosis, and cancer. Upon injury, HSCs activate and are the main origin of myofibroblasts and cancer-associated fibroblasts (CAFs) in liver fibrosis and cancer. Primary liver cancer has a grim prognosis, ranking as the third leading cause of cancer-related deaths worldwide, with hepatocellular carcinoma (HCC) being the predominant type, followed by intrahepatic cholangiocarcinoma (iCCA). Moreover, the liver hosts 35% of all metastatic lesions. The distinct spatial distribution and functional roles of HSCs across these malignancies represent a significant challenge for universal therapeutic strategies, requiring a nuanced and tailored understanding of their contributions. This review examines the heterogeneous roles of HSCs in liver cancer, focusing on their spatial localization, dynamic interactions within the tumor microenvironment (TME), and emerging therapeutic opportunities, including strategies to modulate their activity, and harness their potential as targets for antifibrotic and antitumor interventions.
Collapse
Affiliation(s)
- Laura Sererols-Viñas
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Gemma Garcia-Vicién
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Paloma Ruiz-Blázquez
- University of Barcelona, Barcelona, Spain
- Tissue Remodeling Fibrosis and Cancer Group, Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain
- Institute of Biomedical Research of Barcelona (IDIBAPS), Barcelona, Spain
- CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Ting-Fang Lee
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Youngmin A Lee
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ester Gonzalez-Sanchez
- HepatoBiliary Tumours Lab, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Salamanca, Spain
- Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain
| | - Javier Vaquero
- CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain
- HepatoBiliary Tumours Lab, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Salamanca, Spain
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Anna Moles
- Tissue Remodeling Fibrosis and Cancer Group, Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain
- Institute of Biomedical Research of Barcelona (IDIBAPS), Barcelona, Spain
- CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Aveline Filliol
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Silvia Affò
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
13
|
Sun X, Wang D, Chang S, Yin L, Zhang H, Ji S, Fei H, Jin Y. Development of Cytolytic Iridium-Complexed Octaarginine Peptide Albumin Nanomedicine for Hepatocellular Carcinoma Treatment. Int J Nanomedicine 2025; 20:2395-2409. [PMID: 40027874 PMCID: PMC11871924 DOI: 10.2147/ijn.s502257] [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: 11/11/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
Objective Hepatocellular carcinoma is one of the most challenging malignancies and has high incidence and mortality rates worldwide. Digital subtraction angiography (DSA)-guided hepatic arterial infusion of the standard chemotherapeutic agent oxaliplatin (OXA) has the advantages of both precision and efficacy, making it an important therapeutic strategy for advanced-stage liver cancer. However, patients receiving this treatment still face severe systemic toxicity and poor tolerability of oxaliplatin. Methods In this study, we compared oxaliplatin with novel albumin-formulated oncolytic peptide nanoparticles, Ir-cR8 (abbreviated as iPep), in the treatment of orthotopic liver cancer in a mouse model by intravenous injection and in a rabbit model via DSA-guided hepatic arterial infusion. Results The results showed that intravenous Ir-cR8-BSA-NPs had enhanced inhibitory effects to the growth of H22 ectopic liver tumors in mice and also with reduced toxicity in animals compared to OXA treatment. Specifically, Ir-cR8-BSA-NPs-treated mice showed approximately 92% tumor growth inhibition compared to approximately 88% for OXA. In the rabbit VX2 ectopic hepatocellular carcinoma model, Ir-cR8-BSA-NPs demonstrated significantly stronger inhibition (P<0.01) of tumor size compared to OXA, as assessed by PET/CT imaging, with SUV values decreasing from 5.15±0.46 to 2.52±0.57, compared to OXA-treated group, which decreased from 5.44±0.43 to 3.90±0.24. Furthermore, Ir-cR8- BSA-NPs significantly improved stability by albumin encapsulation and reduced hemolytic toxicity (P<0.001), resulting in improved therapeutic efficacy. Conclusion This study demonstrated the combined advantages of a novel membrane-active oncolytic peptide nanomedicine and precise drug delivery enabled by arterial infusion technology for the interventional treatment of liver cancer.
Collapse
Affiliation(s)
- Xingwei Sun
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, People’s Republic of China
| | - Di Wang
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, People’s Republic of China
| | - Shiwei Chang
- Nanobiomedicine Division, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, People’s Republic of China
| | - Liang Yin
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, People’s Republic of China
| | - Hao Zhang
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, People’s Republic of China
| | - Shuangshuang Ji
- Nanobiomedicine Division, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, People’s Republic of China
| | - Hao Fei
- Nanobiomedicine Division, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, People’s Republic of China
| | - Yong Jin
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, People’s Republic of China
| |
Collapse
|
14
|
Wang X, Wang W, Zeng H, Hu X, Chen F, Shen L, Tao J. Molecular structure of polysaccharide mediated autophagy markers KIF23 and PRC1 proteins and their regulatory role in triple negative cancer through the p53 signaling pathway. Int J Biol Macromol 2025; 291:139155. [PMID: 39725112 DOI: 10.1016/j.ijbiomac.2024.139155] [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: 10/31/2024] [Revised: 12/11/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
As a process of intracellular degradation and recycling of its own components, abnormal regulation of autophagy has been strongly associated with the development of multiple cancer types, including triple-negative breast cancer. The amino acid sequences of KIF23 and PRC1 proteins were analyzed by bioinformatics method, their three-dimensional structures were predicted, and their interactions with polysaccharides were studied by molecular docking technology. The localization and expression patterns of KIF23 and PRC1 in cells were studied by cell biology techniques. By constructing breast cancer cell lines that stably overexpress or knock down KIF23 and PRC1, we evaluated the effect of these proteins on autophagy activity. Finally, molecular biological methods such as Western blot and real-time quantitative PCR were used to detect the expression changes of proteins related to p53 signaling pathway and the levels of autophagy markers such as LC3 and p62, thereby revealing the regulatory effects of KIF23 and PRC1 on autophagy of triple-negative breast cancer cells through p53 signaling pathway. The study found that the KIF23 and PRC1 proteins have complex three-dimensional structures, and their interactions with polysaccharides may affect their function during cell division and autophagy. In triple-negative breast cancer cells, overexpression of KIF23 and PRC1 significantly enhanced autophagy activity, while knockdown of these proteins inhibited autophagy. Further experiments showed that KIF23 and PRC1 regulate the expression of autophagy related proteins by influencing the activity of p53 signaling pathway. Overexpression of KIF23 and PRC1 led to inhibition of the p53 signaling pathway, while knocking down these proteins activated the p53 signaling pathway, which was consistent with reduced autophagy activity.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- Clinical Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Wei Wang
- GCP Center, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Hanling Zeng
- Department of General Surgery, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210031, China
| | - Xinru Hu
- Department of Public Health, School of Medicine and Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Fangyi Chen
- Department of Public Health, School of Medicine and Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li Shen
- Department of General Surgery, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210031, China.
| | - Jing Tao
- Department of General Surgery, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210031, China.
| |
Collapse
|
15
|
Vázquez Salgado AM, Cai C, Lee M, Yin D, Chrystostome ML, Gefre AF, He S, Kieckhaefer JE, Wangensteen KJ. In Vivo CRISPR Activation Screening Reveals Chromosome 1q Genes VPS72, GBA1, and MRPL9 Drive Hepatocellular Carcinoma. Cell Mol Gastroenterol Hepatol 2025; 19:101460. [PMID: 39761726 PMCID: PMC11929076 DOI: 10.1016/j.jcmgh.2025.101460] [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: 06/30/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC) frequently undergoes regional chromosomal amplification, resulting in elevated gene expression levels. We aimed to elucidate the role of these poorly understood genetic changes by using CRISPR activation (CRISPRa) screening in mouse livers to identify which genes within these amplified loci are cancer driver genes. METHODS We used data from The Cancer Genome Atlas to identify that frequently copy number-amplified and up-regulated genes all reside on human chromosomes 1q and 8q. We generated CRISPRa screening transposons that contain oncogenic Myc to drive tumor formation. We conducted CRISPRa screens in vivo in the liver to identify tumor driver genes. We extensively validated the findings in separate mice and performed RNA sequencing analysis to explore mechanisms driving tumorigenesis. RESULTS We targeted genes that frequently undergo amplification in human HCC using an in vivo CRISPRa screening system in mice, which induced extensive liver tumorigenesis. Human chromosome 1q genes Zbtb7b, Vps72, Gba1, and Mrpl9 emerged as drivers of liver tumorigenesis. In human HCC there is a trend in correlation between levels of MRPL9, VPS72, or GBA1 and poor survival. In validation assays, activation of Vps72, Gba1, or Mrpl9 resulted in extensive liver tumorigenesis and decreased survival in mice. RNA sequencing revealed different mechanisms driving HCC, with Mrpl9 activation altering genes functionally related to mitochondrial function, Vps72 levels altering phospholipid metabolism, and Gba1 activation enhancing endosomal-lysosomal activity, all leading to promotion of cellular proliferation. Analysis of human tumor tissues with high levels of MRPL9, VPS72, or GBA1 revealed congruent results, indicating conserved mechanisms driving HCC. CONCLUSIONS This study reveals chromosome 1q genes Vps72, Gba1, and Mrpl9 as drivers of HCC. Future efforts to prevent or treat HCC can focus on these new driver genes.
Collapse
Affiliation(s)
- Alexandra M Vázquez Salgado
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota; Pharmacology Graduate Program, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chunmiao Cai
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Markcus Lee
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Dingzi Yin
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Marie-Lise Chrystostome
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Adrienne F Gefre
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Shirui He
- College of Arts & Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Julia E Kieckhaefer
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kirk J Wangensteen
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
16
|
Wang K, Zhang Y, Si C, Cao Y, Shao P, Zhang P, Wang N, Su G, Qian J, Yang L. Cholesterol: The driving force behind the remodeling of tumor microenvironment in colorectal cancer. Heliyon 2024; 10:e39425. [PMID: 39687190 PMCID: PMC11648115 DOI: 10.1016/j.heliyon.2024.e39425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 12/18/2024] Open
Abstract
Essential membrane components and metabolites with a wide range of biological roles are both produced by cholesterol metabolism. Cell-intrinsic and cell-extrinsic stimuli alter cholesterol metabolism in the tumor microenvironment (TME), which in turn encourages colorectal carcinogenesis. Metabolites produced from cholesterol play intricate roles in promoting the development of colorectal cancer (CRC) and stifling immunological responses. By altering the extracellular matrix of the main tumor, redesigning its immunological environment, and altering its mechanical stiffness, cholesterol can encourage the epithelial-mesenchymal transition of the primary tumor, opening up a pathway for tumor metastasis. Its functions in TME remodeling and tumor prevention have been recently identified. In this review we address the function of cholesterol in TME remodeling and therapeutic techniques designed to block cholesterol metabolism, and discuss how combining these strategies with already available anti-CRC medicines can have combined effects and open up new therapeutic avenues.
Collapse
Affiliation(s)
- Ke Wang
- Department of Colorectal Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yuanyuan Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Chengshuai Si
- Department of Colorectal Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yuepeng Cao
- Department of Colorectal Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Peng Shao
- Department of Colorectal Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Pei Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Nannan Wang
- Department of Colorectal Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Guoqing Su
- Department of Colorectal Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Jinghang Qian
- Department of Colorectal Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Liu Yang
- Department of Colorectal Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| |
Collapse
|
17
|
Zhou P, Qiu C, Zhuang Z, Shi K, Yang Z, Ding Y, Qu H, Xia J. A two-sample Mendelian randomization study reveals the causal effects of statin medication on gut microbiota abundance in the European population. Front Genet 2024; 15:1380830. [PMID: 39734574 PMCID: PMC11674602 DOI: 10.3389/fgene.2024.1380830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 11/14/2024] [Indexed: 12/31/2024] Open
Abstract
Background Observational studies have reported changes in gut microbiota abundance caused by long-term statin medication therapy. However, the causal relation between statin medication and gut microbiota subsets based on genetic variants remains unclear. Methods We used genome-wide association study (GWAS) data on statin medication from the FinnGen database and gut microbiota abundance GWAS data from the IEU OpenGWAS project. A Mendelian randomization (MR) analysis was conducted to evaluate the causal effect of statin medication on gut microbiota abundance using the inverse variance weighting (IVW) method, MR-Egger regression, and weighted median approach. Meanwhile, heterogeneity and pleiotropy analyses were also undertaken in this study. Results Statin medication was negatively correlated with five species of gut microbiota abundance: Parabacteroides (BetaIVW = -0.2745, 95% CI = (-0.4422, -0.1068), and P IVW = 0.0013), Ruminococcaceae UCG-009 (BetaIVW = -0.1904, 95% CI = (-0.3255, -0.0553), and P IVW = 0.0057), Coprococcus 1 (BetaIVW = -0.1212, 95% CI = (-0.2194, -0.0231), and P IVW = 0.0154), Ruminococcaceae UCG-010 (BetaIVW = -0.1149, 95% CI = (-0.2238, -0.0060), and P IVW = 0.0385), and Veillonellaceae (BetaIVW = -0.0970, 95% CI = (-0.2238, 0.0060), and P IVW = 0.0400) and positively correlated with one species of gut microbiota: Desulfovibrio (BetaIVW = 0.2452, 95% CI = (0.0299, 0.4606), and P IVW = 0.0255). In addition, no significant heterogeneity or pleiotropy was detected in the abovementioned gut microbiota. Conclusion This Mendelian randomization analysis indicates a causal relationship between statin medication and six gut microbiota species. These findings may provide new strategies for health monitoring in populations taking long-term statin medications.
Collapse
Affiliation(s)
- Peng Zhou
- Department of General Surgery, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center, School of Medicine, Jiangnan University, Wuxi, China
| | - Chen Qiu
- Department of General Surgery, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Zequn Zhuang
- Department of Hepatobiliary Surgery, Jinjiang Municipal Hospital (Shanghai Sixth People’s Hospital Fujian), Quanzhou, Fujian, China
| | - Kaihang Shi
- Department of Hepatobiliary Surgery, The Affiliated Yixing Hospital of Jiangsu University, Wuxi, China
| | - Zhihui Yang
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center, School of Medicine, Jiangnan University, Wuxi, China
| | - Yuyan Ding
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center, School of Medicine, Jiangnan University, Wuxi, China
| | - Huiheng Qu
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center, School of Medicine, Jiangnan University, Wuxi, China
| | - Jiazeng Xia
- Department of General Surgery, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center, School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
18
|
Nguyen MT, Lee GJ, Kim B, Kim HJ, Tak J, Park MK, Kim EJ, Kang GJ, Rho SB, Lee H, Lee K, Kim SG, Lee CH. Penfluridol suppresses MYC-driven ANLN expression and liver cancer progression by disrupting the KEAP1-NRF2 interaction. Pharmacol Res 2024; 210:107512. [PMID: 39643070 DOI: 10.1016/j.phrs.2024.107512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/05/2024] [Accepted: 11/16/2024] [Indexed: 12/09/2024]
Abstract
Hepatocellular carcinoma (HCC) comprises the majority of primary liver cancers and possesses a low 5-year survival rate when in the advanced stages. Anillin (ANLN), a key player in cell growth and cytokinesis, is implicated in HCC development. Currently, no treatment agents are known to suppress ANLN. Analysis of The Cancer Genome Atlas data showed that high ANLN expression is associated with poor prognosis and survival in HCC patients. ANLN knockdown was shown to inhibit proliferation, cell cycle progression, and PD-L1 expression in liver cancer cells. The antipsychotic drug penfluridol was identified to suppress ANLN expression in the Connectivity Map analysis. Penfluridol downregulated ANLN at both the mRNA and protein levels, leading to G2/M cell cycle arrest and reduced colony formation in liver cancer cells. Mechanistically, penfluridol inhibited the transcription factor MYC from binding to an E-box motif in the ANLN promoter. This process was mediated by penfluridol-induced upregulation of NRF2, which competitively bound and sequestered MYC away from the ANLN promoter. Penfluridol inhibited the interaction between NRF2 and KEAP1, increasing NRF2. In a syngeneic mouse model, penfluridol suppressed liver tumour growth accompanied by increased NRF2 and decreased MYC and ANLN expression. These findings suggest penfluridol can be applied as the first ANLN blocker to modulate the MYC/NRF2/KEAP1 axis.
Collapse
Affiliation(s)
- Minh Tuan Nguyen
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Gi Jeong Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Boram Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Hyun Ji Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Jihoon Tak
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Mi Kyung Park
- Department of Bio-Healthcare, Hwasung Medi-Science University, Hwaseong-si 18274, Republic of Korea
| | - Eun Ji Kim
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Gyeoung Jin Kang
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Seung Bae Rho
- National Cancer Center, Goyang 10408, Republic of Korea
| | - Ho Lee
- National Cancer Center, Goyang 10408, Republic of Korea
| | - Kyung Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Sang Geon Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Chang Hoon Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea.
| |
Collapse
|
19
|
Zhang L, Gao M, Wu Y, Liu H, Zhuang X, Zhou Y, Song Q, Bi S, Zhang W, Cui Y. MST1 interactomes profiling across cell death in esophageal squamous cell carcinoma. MEDICAL REVIEW (2021) 2024; 4:531-543. [PMID: 39664081 PMCID: PMC11629308 DOI: 10.1515/mr-2024-0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/27/2024] [Indexed: 12/13/2024]
Abstract
Objectives Resistance to apoptosis in esophageal squamous cell carcinoma (ESCC) constitutes a significant impediment to treatment efficacy. Exploring alternative cell death pathways and their regulatory factors beyond apoptosis is crucial for overcoming drug resistance and enhancing therapeutic outcomes in ESCC. Methods Mammalian Ste 20-like kinase 1 (MST1) is implicated in regulating various cell deaths, including apoptosis, autophagy, and pyroptosis. Employing enhanced ascorbate peroxidase 2 (APEX2) proximity labeling coupled with immunoprecipitation-mass spectrometry (IP-MS), we elucidated the interactomes of MST1 across these three cell death paradigms. Results Proteomic profiling unveiled the functional roles and subcellular localization of MST1 and its interacting proteins during normal proliferation and various cell death processes. Notably, MST1 exhibited an expanded interactome during cell death compared to normal proliferation and chromosome remodeling functions consistently. In apoptosis, there was a notable increase of mitosis-associated proteins such as INCENP, ANLN, KIF23, SHCBP1 and SUPT16H, which interacted with MST1, alongside decreased expression of the pre-apoptotic protein STK3. During autophagy, the bindings of DNA repair-related proteins CBX8 and m6A reader YTHDC1 to MST1 were enhanced. In pyroptosis, LRRFIP2 and FLII which can inhibit pyroptosis increasingly binding to MST1. Conclusions Our findings delineate potential mechanisms through which MST1 and its interactomes regulate cell death, paving the way for further investigation to validate and consolidate these observations.
Collapse
Affiliation(s)
- Li Zhang
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Mingwei Gao
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yueguang Wu
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Huijuan Liu
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Xuehan Zhuang
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Yan Zhou
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Qiqin Song
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Shanshan Bi
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Weimin Zhang
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Yongping Cui
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| |
Collapse
|
20
|
Lehrich BM, Tao J, Liu S, Hirsch TZ, Yasaka TM, Cao C, Delgado ER, Guan X, Lu S, Pan L, Liu Y, Singh S, Poddar M, Bell A, Singhi AD, Zucman-Rossi J, Wang Y, Monga SP. Development of mutated β-catenin gene signature to identify CTNNB1 mutations from whole and spatial transcriptomic data in patients with HCC. JHEP Rep 2024; 6:101186. [PMID: 39583094 PMCID: PMC11582745 DOI: 10.1016/j.jhepr.2024.101186] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 11/26/2024] Open
Abstract
Background & Aims Patients with β-catenin (encoded by CTNNB1)-mutated hepatocellular carcinoma (HCC) demonstrate heterogenous responses to first-line immune checkpoint inhibitors (ICIs). Precision-medicine based treatments for this subclass are currently in clinical development. Here, we report derivation of the Mutated β-catenin Gene Signature (MBGS) to predict CTNNB1-mutational status in patients with HCC for future application in personalized medicine treatment regimens. Methods Co-expression of mutant-Nrf2 and hMet ± mutant-β-catenin in murine livers in mice led to HCC development. The MBGS was derived using bulk RNA-seq and intersectional transcriptomic analysis of β-catenin-mutated and non-mutated HCC models. Integrated RNA/whole-exome-sequencing and spatial transcriptomic data from multiple cohorts of patients with HCC was assessed to address the ability of MBGS to detect CTNNB1 mutation, the tumor immune microenvironment, and/or predict therapeutic responses. Results Bulk RNA-seq comparing HCC specimens in mutant β-catenin-Nrf2, β-catenin-Met and β-catenin-Nrf2-Met to Nrf2-Met HCC model yielded 95 common upregulated genes. In The Cancer Genome Atlas (TCGA)-LIHC dataset, differential gene expression analysis with false discovery rate (FDR) = 0.05 and log2(fold change) >1.5 on the 95 common genes comparing CTNNB1-mutated vs. wild-type patients narrowed the gene panel to a 13-gene MBGS. MBGS predicted CTNNB1-mutations in TCGA (n = 374) and French (n = 398) patient cohorts with AUCs of 0.90 and 0.94, respectively. Additionally, a higher MBGS expression score was associated with lack of significant improvement in overall survival or progression-free survival in the atezolizumab-bevacizumab arm vs. the sorafenib arm in the IMbrave150 cohort. MBGS performed comparable or superior to other CTNNB1-mutant classifiers. MBGS overlapped with Hoshida S3, Boyault G5/G6, and Chiang CTNNB1 subclass tumors in TCGA and in HCC spatial transcriptomic datasets visually depicting these tumors to be situated in an immune excluded tumor microenvironment. Conclusions MBGS will aid in patient stratification to guide precision medicine therapeutics for CTNNB1-mutated HCC subclass as a companion diagnostic, as anti-β-catenin therapies become available. Impact and implications As precision medicine for liver cancer treatment becomes a reality, diagnostic tools are needed to help classify patients into groups for the best treatment choices. We have developed a molecular signature that could serve as a companion diagnostic and uses bulk or spatial transcriptomic data to identify a unique subclass of liver tumors. This subgroup of liver cancer patients derive limited benefit from the current standard of care and are expected to benefit from specialized directed therapies that are on the horizon.
Collapse
Affiliation(s)
- Brandon M. Lehrich
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Junyan Tao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Silvia Liu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Theo Z. Hirsch
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, Paris, France
- Institut du Cancer Paris CARPEM, AP-HP, Department of Oncology, Hopital Européen Georges Pompidou, Paris, France
| | - Tyler M. Yasaka
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Catherine Cao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Evan R. Delgado
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Xiangnan Guan
- Translational Medicine, Genentech Inc., San Francisco, CA, USA
| | - Shan Lu
- Translational Medicine, Genentech Inc., San Francisco, CA, USA
| | - Long Pan
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, Paris, France
- Institut du Cancer Paris CARPEM, AP-HP, Department of Oncology, Hopital Européen Georges Pompidou, Paris, France
| | - Yuqing Liu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sucha Singh
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Minakshi Poddar
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Aaron Bell
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Aatur D. Singhi
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, Paris, France
- Institut du Cancer Paris CARPEM, AP-HP, Department of Oncology, Hopital Européen Georges Pompidou, Paris, France
| | - Yulei Wang
- Translational Medicine, Genentech Inc., San Francisco, CA, USA
| | - Satdarshan P. Monga
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
21
|
Hu S, Gan M, Wei Z, Shang P, Song L, Feng J, Chen L, Niu L, Wang Y, Zhang S, Shen L, Zhu L, Zhao Y. Identification of host factors for livestock and poultry viruses: genome-wide screening technology based on the CRISPR system. Front Microbiol 2024; 15:1498641. [PMID: 39640855 PMCID: PMC11619636 DOI: 10.3389/fmicb.2024.1498641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Genome-wide CRISPR library screening technology is a gene function research tool developed based on the CRISPR/Cas9 gene-editing system. The clustered regularly interspaced short palindromic repeats/CRISPR-associated genes (CRISPR/Cas) system, considered the third generation of gene editing after zinc finger nucleases (ZFN) and transcription activator-like effector nucleases (TALEN), is widely used for screening various viral host factors. CRISPR libraries are classified into three main categories based on the different functions of Cas9 enzymes: CRISPR knockout (CRISPR KO) library screening, CRISPR transcriptional activation (CRISPRa) library screening, and CRISPR transcriptional interference (CRISPRi) library screening. Recently, genome-wide CRISPR library screening technology has been used to identify host factors that interact with viruses at various stages, including adsorption, endocytosis, and replication. By specifically modulating the expression of these host factors, it becomes possible to cultivate disease-resistant varieties, establish disease models, and design and develop vaccines, among other applications. This review provides an overview of the development and technical processes of genome-wide CRISPR library screening, as well as its applications in identifying viral host factors in livestock and poultry.
Collapse
Affiliation(s)
- Shijie Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Mailin Gan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Ziang Wei
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Pan Shang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Lei Song
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Jinkang Feng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Lei Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Lili Niu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Ya’an, China
| | - Shunhua Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Linyuan Shen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Ya’an, China
| | - Li Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Ya’an, China
| | - Ye Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Ya’an, China
| |
Collapse
|
22
|
Zhang H, Han B, Tian S, Gong Y, Liu L. ZNF740 facilitates the malignant progression of hepatocellular carcinoma via the METTL3/HIF‑1A signaling axis. Int J Oncol 2024; 65:105. [PMID: 39301659 PMCID: PMC11436261 DOI: 10.3892/ijo.2024.5693] [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: 01/25/2024] [Accepted: 07/11/2024] [Indexed: 09/22/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer‑related death, and efficient treatments to facilitate recovery and enhance long‑term outcomes are lacking. Zinc finger proteins (ZNFs), known as the largest group of transcription factors, have gained interest for their roles in HCC by stimulating the transcription of well‑known tumor‑causing genes. However, the specific roles and molecular mechanisms of ZNF740 in HCC remain unknown. The present study performed bioinformatics analysis and RNA‑sequencing analysis of differentially expressed genes in HCC, detected ZNF740 expression levels in HCC using reverse transcription‑quantitative PCR, western blotting and immunohistochemistry, and explored the effects of ZNF740 on the progression of liver cancer in vitro and in vivo using cellular functionality assays and cell‑derived xenografts. In addition, a dual‑luciferase reporter assay was performed to analyze the binding of ZNF740 with the METTL3 promoter. Furthermore, cell functionality experiments were performed to analyze whether ZNF740 promotes the proliferation of liver cancer cells in a METTL3‑dependent manner. Bioinformatics and immunoprecipitation assays were further used to analyze the molecular mechanism of ZNF740 in liver cancer. The present study demonstrated that ZNF740 expression was upregulated in HCC. Mechanistically, overexpressed ZNF740 interacted with the methyltransferase‑like 3 (METTL3) promoter and increased METTL3 expression, leading to the stabilization of hypoxia‑inducible factor‑1A (HIF1A) mRNA in an N6‑methyladenosine/YTH N6‑methyladenosine RNA‑binding protein 1‑dependent manner. Eventually, the ZNF740/METTL3/HIF1A signaling axis may facilitate the proliferation, invasion and metastasis of liver cancer via METTL3/HIF‑1A signaling. The present findings revealed the important role of ZNF740 and suggested a potential therapeutic approach that might improve clinical therapies for liver cancer.
Collapse
Affiliation(s)
- Hao Zhang
- College of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Bing Han
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| | - She Tian
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Yongjun Gong
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Li Liu
- School of Public Health, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| |
Collapse
|
23
|
Wang J, Ying L, Xiong H, Zhou DR, Wang YX, Che HL, Zhong ZF, Wu GS, Ge YJ. Comprehensive analysis of stearoyl-coenzyme A desaturase in prostate adenocarcinoma: insights into gene expression, immune microenvironment and tumor progression. Front Immunol 2024; 15:1460915. [PMID: 39351232 PMCID: PMC11439642 DOI: 10.3389/fimmu.2024.1460915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
Prostate adenocarcinoma (PRAD) is a prevalent global malignancy which depends more on lipid metabolism for tumor progression compared to other cancer types. Although Stearoyl-coenzyme A desaturase (SCD) is documented to regulate lipid metabolism in multiple cancers, landscape analysis of its implications in PRAD are still missing at present. Here, we conducted an analysis of diverse cancer datasets revealing elevated SCD expression in the PRAD cohort at both mRNA and protein levels. Interestingly, the elevated expression was associated with SCD promoter hypermethylation and genetic alterations, notably the L134V mutation. Integration of comprehensive tumor immunological and genomic data revealed a robust positive correlation between SCD expression levels and the abundance of CD8+ T cells and macrophages. Further analyses identified significant associations between SCD expression and various immune markers in tumor microenvironment. Single-cell transcriptomic profiling unveiled differential SCD expression patterns across distinct cell types within the prostate tumor microenvironment. The Gene Ontology and Kyoto Encyclopedia of Genes and Genome analyses showed that SCD enriched pathways were primarily related to lipid biosynthesis, cholesterol biosynthesis, endoplasmic reticulum membrane functions, and various metabolic pathways. Gene Set Enrichment Analysis highlighted the involvement of elevated SCD expression in crucial cellular processes, including the cell cycle and biosynthesis of cofactors pathways. In functional studies, SCD overexpression promoted the proliferation, metastasis and invasion of prostate cancer cells, whereas downregulation inhibits these processes. This study provides comprehensive insights into the multifaceted roles of SCD in PRAD pathogenesis, underscoring its potential as both a therapeutic target and prognostic biomarker.
Collapse
Affiliation(s)
- Jie Wang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Liang Ying
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - He Xiong
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Duan-Rui Zhou
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yi-Xuan Wang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Hui-Lian Che
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhang-Feng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| | - Guo-Sheng Wu
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yun-Jun Ge
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
24
|
Chen Y, Wong CCL. The mechanistic insights behind the anticancer effects of statins in liver cancer. Hepatol Commun 2024; 8:e0519. [PMID: 39225688 PMCID: PMC11371310 DOI: 10.1097/hc9.0000000000000519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Affiliation(s)
- Yiling Chen
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Carmen Chak-Lui Wong
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong, China
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
25
|
Zhou W, Ye F, Yang G, Liu C, Pan Z, Zhang C, Liu H. YAP-based nomogram predicts poor prognosis in patients with hepatocellular carcinoma after curative surgery. J Gastrointest Oncol 2024; 15:1712-1722. [PMID: 39279983 PMCID: PMC11399831 DOI: 10.21037/jgo-24-36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 06/14/2024] [Indexed: 09/18/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC) ranks prominently in cancer-related mortality globally. Surgery remains the main therapeutic option for the treatment of HCC, but high post-operative recurrence rate makes prognostic prediction challenging. The quest for a reliable model to predict HCC recurrence continues to enhance prognosis. We aim to develop a nomogram with multiple factors to accurately estimate the risk of post-operative recurrence in patients with HCC. Methods A single-center retrospective study on 262 patients who underwent partial hepatectomy for HCC at the Eastern Hepatobiliary Surgery Hospital from May 2010 to April 2013 was conducted where immunohistochemistry assessed Yes-associated protein (YAP) expression in HCC. In the training cohort, a nomogram that incorporated YAP expression and clinicopathological features was constructed to predict 2-, 3-, and 5-year recurrence-free survival (RFS). The performance of the nomogram was assessed with respect to discrimination calibration, and clinical usefulness with external validation. Results A total of 262 patients who underwent partial hepatectomy for HCC at the Eastern Hepatobiliary Surgery Hospital were included in our study. HCC patients with high YAP expression exhibited significantly higher recurrence and reduced overall survival (OS) rates compared to those with low YAP expression (P<0.001). YAP was significantly associated with alpha-fetoprotein (AFP) (P=0.03), microvascular invasion (MVI) (P<0.001), and tumor differentiation grade (P<0.001). In the training cohort, factors like YAP expression, hepatitis B surface antigen (HBsAg), hepatitis B virus deoxyribonucleic acid (HBV-DNA), Child-Pugh stage, tumor size, MVI, and tumor differentiation were identified as key elements for the predictive model. Two YAP-centric Nomograms were developed, with one focused on predicting postoperative OS and the other on RFS. The calibration curve further confirmed the model's accuracy in the training cohort. The validation cohort confirmed the model's predictive accuracy. Conclusions The proposed nomogram combining the YAP, a predictor of HCC progression, and clinical features achieved more-accurate prognostic prediction for patients with HCC after partial hepatectomy, which may help clinicians implement more appropriate interventions.
Collapse
Affiliation(s)
- Wenxuan Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital (Second Military Medical University), Naval Medical University, Shanghai, China
| | - Feiyang Ye
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital (Second Military Medical University), Naval Medical University, Shanghai, China
| | - Gaowei Yang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital (Second Military Medical University), Naval Medical University, Shanghai, China
| | - Chenghu Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital (Second Military Medical University), Naval Medical University, Shanghai, China
| | - Zeya Pan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital (Second Military Medical University), Naval Medical University, Shanghai, China
| | - Chengjing Zhang
- Department of Nutrition, Eastern Hepatobiliary Surgery Hospital (Second Military Medical University), Naval Medical University, Shanghai, China
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital (Second Military Medical University), Naval Medical University, Shanghai, China
| |
Collapse
|
26
|
Li T, Li S, Kang Y, Zhou J, Yi M. Harnessing the evolving CRISPR/Cas9 for precision oncology. J Transl Med 2024; 22:749. [PMID: 39118151 PMCID: PMC11312220 DOI: 10.1186/s12967-024-05570-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024] Open
Abstract
The Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)/Cas9 system, a groundbreaking innovation in genetic engineering, has revolutionized our approach to surmounting complex diseases, culminating in CASGEVY™ approved for sickle cell anemia. Derived from a microbial immune defense mechanism, CRISPR/Cas9, characterized as precision, maneuverability and universality in gene editing, has been harnessed as a versatile tool for precisely manipulating DNA in mammals. In the process of applying it to practice, the consecutive exploitation of novel orthologs and variants never ceases. It's conducive to understanding the essentialities of diseases, particularly cancer, which is crucial for diagnosis, prevention, and treatment. CRISPR/Cas9 is used not only to investigate tumorous genes functioning but also to model disparate cancers, providing valuable insights into tumor biology, resistance, and immune evasion. Upon cancer therapy, CRISPR/Cas9 is instrumental in developing individual and precise cancer therapies that can selectively activate or deactivate genes within tumor cells, aiming to cripple tumor growth and invasion and sensitize cancer cells to treatments. Furthermore, it facilitates the development of innovative treatments, enhancing the targeting efficiency of reprogrammed immune cells, exemplified by advancements in CAR-T regimen. Beyond therapy, it is a potent tool for screening susceptible genes, offering the possibility of intervening before the tumor initiative or progresses. However, despite its vast potential, the application of CRISPR/Cas9 in cancer research and therapy is accompanied by significant efficacy, efficiency, technical, and safety considerations. Escalating technology innovations are warranted to address these issues. The CRISPR/Cas9 system is revolutionizing cancer research and treatment, opening up new avenues for advancements in our understanding and management of cancers. The integration of this evolving technology into clinical practice promises a new era of precision oncology, with targeted, personalized, and potentially curative therapies for cancer patients.
Collapse
Affiliation(s)
- Tianye Li
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, People's Republic of China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, 310000, People's Republic of China
| | - Shuiquan Li
- Department of Rehabilitation and Traditional Chinese Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, People's Republic of China
| | - Yue Kang
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, People's Republic of China.
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, 310000, People's Republic of China.
| | - Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|
27
|
Kim J, Seki E. Unveiling the cancer risk nexus of the steatotic liver. Trends Endocrinol Metab 2024; 35:708-719. [PMID: 38531699 PMCID: PMC11321945 DOI: 10.1016/j.tem.2024.02.017] [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: 01/09/2024] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024]
Abstract
Steatotic liver, characterized by the accumulation of fat in the liver, poses significant health risks including metabolic dysfunction-associated steatotic liver disease (MASLD) and an elevated risk of primary liver cancer. Emerging evidence indicates a robust association between steatotic liver and increased susceptibility to extrahepatic primary cancers and their metastases. The deposition of fat induces dynamic changes in hepatic microenvironments, thereby fostering inflammation and immune responses that enhance liver metastasis from extrahepatic primary cancers. This review explores the impact of steatotic liver on hepatic carcinogenesis and metastasis from extrahepatic cancers, with a specific focus on hepatocyte-derived factors and the immune microenvironment. By emphasizing novel conclusions, this article underscores the timely relevance of understanding these intricate connections.
Collapse
Affiliation(s)
- Jieun Kim
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
28
|
Li N, Wang G, Guo M, Zhu N, Yu W. The mechanism and clinical application of farnesyl diphosphate farnesyltransferase 1 in cancer metabolism. Biochem Biophys Res Commun 2024; 719:150046. [PMID: 38749088 DOI: 10.1016/j.bbrc.2024.150046] [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: 03/10/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 06/05/2024]
Abstract
Cancer poses a significant risk to human well-being. Among the crucial characteristics of cancer is metabolic reprogramming. To meet the relentless metabolic needs, cancer cells enhance cholesterol metabolism within the adverse tumor microenvironment. Reprograming cholesterol metabolism includes a series of modifications in the synthesis, absorption, esterification, and metabolites associated with cholesterol. These adjustments have a strong correlation with the proliferation, invasion, metastasis, and other characteristics of malignant tumors. FDFT1, also known as farnesyl diphosphate farnesyltransferase 1, is an enzyme crucial in the process of cholesterol biosynthesis. Its significant involvement in tumor metabolism has garnered considerable interest. The significance of FDFT1 in cancer metabolism cannot be overstated, as it actively interacts with cancer cells. This paper aims to analyze and consolidate the mechanism of FDFT1 in cancer metabolism and explore its clinical application. The goal is to contribute new strategies and targets for the prevention and treatment of cancer metabolism.
Collapse
Affiliation(s)
- Nanxin Li
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Guojuan Wang
- Department of Oncology, Affiliated Hospital of Jiangxi University of Chinese Medicine, No.445, Bayi Avenue, Nanchang, 330006, China.
| | - Min Guo
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Naicheng Zhu
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Wenyan Yu
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| |
Collapse
|
29
|
Lv X, Lan G, Zhu L, Guo Q. Breaking the Barriers of Therapy Resistance: Harnessing Ferroptosis for Effective Hepatocellular Carcinoma Therapy. J Hepatocell Carcinoma 2024; 11:1265-1278. [PMID: 38974015 PMCID: PMC11227329 DOI: 10.2147/jhc.s469449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/11/2024] [Indexed: 07/09/2024] Open
Abstract
Ferroptosis is a type of cell death that relies on iron and is distinguished by the occurrence of lipid peroxidation and the buildup of reactive oxygen species. Ferroptosis has been demonstrated to have a significant impact on the advancement and resistance to treatment of hepatocellular carcinoma (HCC), thereby highlighting its potential as a viable therapeutic target. Ferroptosis was observed in HCC tissues in contrast to normal liver tissue. The inhibition of ferroptosis has been found to increase the viability of HCC cells and decrease their susceptibility to various anticancer therapies, including chemotherapy, radiotherapy, and immune checkpoint blockade. The administration of drugs that directly modulate ferroptosis regulators or induce excessive production of lipid-reactive oxygen species has demonstrated the potential to enhance the responsiveness of drug-resistant HCC cells to treatment. However, the precise mechanism underlying this phenomenon remains ambiguous. This review presents a comprehensive overview of the crucial role played by ferroptosis in enhancing the efficacy of treatment for hepatocellular carcinoma (HCC). The main aim of this study is to examine the feasibility of utilizing ferroptosis as a therapeutic approach to improve the efficacy of HCC treatment and overcome drug resistance.
Collapse
Affiliation(s)
- Xianmei Lv
- Department of Radiotherapy, Jinhua People’s Hospital, Jinhua, Zhejiang, 321000, People’s Republic of China
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, People’s Republic of China
| | - Gaochen Lan
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, People’s Republic of China
| | - Lujian Zhu
- Department of Medical Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, People’s Republic of China
| | - Qiusheng Guo
- Department of Radiotherapy, Jinhua People’s Hospital, Jinhua, Zhejiang, 321000, People’s Republic of China
| |
Collapse
|
30
|
Xie Q, Zeng Y, Zhang X, Yu F. The significance of lipid metabolism reprogramming of tumor-associated macrophages in hepatocellular carcinoma. Cancer Immunol Immunother 2024; 73:171. [PMID: 38954021 PMCID: PMC11220057 DOI: 10.1007/s00262-024-03748-9] [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: 02/15/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024]
Abstract
In the intricate landscape of the tumor microenvironment, tumor-associated macrophages (TAMs) emerge as a ubiquitous cellular component that profoundly affects the oncogenic process. The microenvironment of hepatocellular carcinoma (HCC) is characterized by a pronounced infiltration of TAMs, underscoring their pivotal role in modulating the trajectory of the disease. Amidst the evolving therapeutic paradigms for HCC, the strategic reprogramming of metabolic pathways presents a promising avenue for intervention, garnering escalating interest within the scientific community. Previous investigations have predominantly focused on elucidating the mechanisms of metabolic reprogramming in cancer cells without paying sufficient attention to understanding how TAM metabolic reprogramming, particularly lipid metabolism, affects the progression of HCC. In this review article, we intend to elucidate how TAMs exert their regulatory effects via diverse pathways such as E2F1-E2F2-CPT2, LKB1-AMPK, and mTORC1-SREBP, and discuss correlations of TAMs with these processes and the characteristics of relevant pathways in HCC progression by consolidating various studies on TAM lipid uptake, storage, synthesis, and catabolism. It is our hope that our summary could delineate the impact of specific mechanisms underlying TAM lipid metabolic reprogramming on HCC progression and provide useful information for future research on HCC and the development of new treatment strategies.
Collapse
Affiliation(s)
- Qingjian Xie
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuan Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangting Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Fujun Yu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
31
|
Moeckel C, Mouratidis I, Chantzi N, Uzun Y, Georgakopoulos-Soares I. Advances in computational and experimental approaches for deciphering transcriptional regulatory networks: Understanding the roles of cis-regulatory elements is essential, and recent research utilizing MPRAs, STARR-seq, CRISPR-Cas9, and machine learning has yielded valuable insights. Bioessays 2024; 46:e2300210. [PMID: 38715516 PMCID: PMC11444527 DOI: 10.1002/bies.202300210] [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: 10/31/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024]
Abstract
Understanding the influence of cis-regulatory elements on gene regulation poses numerous challenges given complexities stemming from variations in transcription factor (TF) binding, chromatin accessibility, structural constraints, and cell-type differences. This review discusses the role of gene regulatory networks in enhancing understanding of transcriptional regulation and covers construction methods ranging from expression-based approaches to supervised machine learning. Additionally, key experimental methods, including MPRAs and CRISPR-Cas9-based screening, which have significantly contributed to understanding TF binding preferences and cis-regulatory element functions, are explored. Lastly, the potential of machine learning and artificial intelligence to unravel cis-regulatory logic is analyzed. These computational advances have far-reaching implications for precision medicine, therapeutic target discovery, and the study of genetic variations in health and disease.
Collapse
Affiliation(s)
- Camille Moeckel
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Ioannis Mouratidis
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Nikol Chantzi
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Yasin Uzun
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
32
|
Liu X, Xie X, Li Q, Xie X, Xiong M, Han W, Xie W. KIF23 promotes cervical cancer progression via inhibiting NLRP3-mediated pyroptosis. FASEB J 2024; 38:e23685. [PMID: 38780518 DOI: 10.1096/fj.202400281r] [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: 02/05/2024] [Revised: 04/25/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Cervical cancer (CC), closely linked to persistent human papillomavirus infection, represents a major health problem for women worldwide. The objective of this study is to elucidate KIF23's role in the development of CC and its regulatory mechanism. METHODS The bioinformatics methods were utilized to extract pyroptosis-associated differentially expressed genes (DEGs) and pivot genes from the GSE9750 and GSE63678 datasets, followed by immune infiltration analysis and quantification of these genes' expression. The effects of kinesin family member 23 (KIF23) were verified through functional experiments in vitro and a mouse xenograft model. The NLPR3 activator, nigericin, was applied for further analyzing the potential regulatory mechanism of KIF23 in CC. RESULTS A total of 8 pyroptosis-related DEGs were screened out, among which 4 candidate core genes were identified as candidate hub genes and confirmed upregulation in CC tissues and cells. These genes respectively showed a positive correlation with the infiltration of distinct immune cells or tumor purity. Downregulation of KIF23 could suppress the proliferation, migration, and invasion abilities in CC cells and tumorigenesis through enhancing pyroptosis. Conversely, KIF23 overexpression accelerated the malignant phenotypes of CC cells and inhibited pyroptosis activation, which was blocked by nigericin treatment. CONCLUSIONS KIF23 may play an oncogenic role in CC progression via inhibition of the NLRP3-mediated pyroptosis pathway.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Xiaoqing Xie
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Qiulian Li
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Xiaohong Xie
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Min Xiong
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Wenling Han
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Wei Xie
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| |
Collapse
|
33
|
Zhao K, Li X, Feng Y, Wang J, Yao W. The role of kinesin family members in hepatobiliary carcinomas: from bench to bedside. Biomark Res 2024; 12:30. [PMID: 38433242 PMCID: PMC10910842 DOI: 10.1186/s40364-024-00559-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/03/2024] [Indexed: 03/05/2024] Open
Abstract
As a major component of the digestive system malignancies, tumors originating from the hepatic and biliary ducts seriously endanger public health. The kinesins (KIFs) are molecular motors that enable the microtubule-dependent intracellular trafficking necessary for mitosis and meiosis. Normally, the stability of KIFs is essential to maintain cell proliferation and genetic homeostasis. However, aberrant KIFs activity may destroy this dynamic stability, leading to uncontrolled cell division and tumor initiation. In this work, we have made an integral summarization of the specific roles of KIFs in hepatocellular and biliary duct carcinogenesis, referring to aberrant signal transduction and the potential for prognostic evaluation. Additionally, current clinical applications of KIFs-targeted inhibitors have also been discussed, including their efficacy advantages, relationship with drug sensitivity or resistance, the feasibility of combination chemotherapy or other targeted agents, as well as the corresponding clinical trials. In conclusion, the abnormally activated KIFs participate in the regulation of tumor progression via a diverse range of mechanisms and are closely associated with tumor prognosis. Meanwhile, KIFs-aimed inhibitors also carry out a promising tumor-targeted therapeutic strategy that deserves to be further investigated in hepatobiliary carcinoma (HBC).
Collapse
Affiliation(s)
- Kai Zhao
- Department of Biliary and Pancreatic Surgery, Cancer Research Center Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Xiangyu Li
- Department of Thoracic Surgery Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Yunxiang Feng
- Department of Biliary and Pancreatic Surgery, Cancer Research Center Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Jianming Wang
- Department of Biliary and Pancreatic Surgery, Cancer Research Center Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China.
- Affiliated Tianyou Hospital, Wuhan University of Science & Technology, 430064, Wuhan, China.
| | - Wei Yao
- Department of Oncology Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China.
| |
Collapse
|
34
|
Jia L, Tian H, Sun S, Hao X, Wen Y. EID3 inhibits the osteogenic differentiation of periodontal ligament stem cells and mediates the signal transduction of TAZ-EID3-AKT/MTOR/ERK. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119662. [PMID: 38216090 DOI: 10.1016/j.bbamcr.2024.119662] [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: 06/05/2023] [Revised: 12/16/2023] [Accepted: 01/03/2024] [Indexed: 01/14/2024]
Abstract
Exploring the molecular mechanisms of cell behaviors is beneficial for promoting periodontal ligament stem cell (PDLSC)-mediated tissue regeneration. This study intends to explore the regulatory effects of EID3 on cell proliferation, apoptosis, and osteogenic differentiation and to preliminarily explore the regulatory mechanism of EID3. Here, EID3 was overexpressed or knocked down in PDLSCs by recombinant lentivirus. Then, cell proliferation activity was analyzed by colony-forming assay, EdU assay, and cell cycle assay. Cell apoptosis was detected by flow cytometry. The osteo-differentiation potential was analyzed using ALP activity assay, ALP staining, alizarin red staining, and mRNA and protein assay of osteo-differentiation related genes. The results showed that when EID3 was knocked down, the proliferation activity and osteogenic differentiation potential of PDLSCs decreased, while they increased when EID3 was overexpressed. The cell apoptosis rate decreased in PDLSCs with EID3 knockdown but increased in PDLSCs with EID3 overexpression. Moreover, EID3 inhibited the transduction of the AKT/MTOR and ERK signaling pathway. In addition, TAZ negatively regulated the expression of EID3, and the overexpression of EID3 partially reversed the promotive effects of TAZ on the osteogenic differentiation of PDLSCs. Taken together, EID3 inhibits the proliferation and osteogenic differentiation while promoting the apoptosis of PDLSCs. EID3 inhibits the transduction of the AKT/MTOR and ERK signaling pathways and mediates the regulatory effect of TAZ on PDLSC osteogenic differentiation.
Collapse
Affiliation(s)
- Linglu Jia
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Shandong, China
| | - Hui Tian
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Stomatology, Rizhao People's Hospital, Rizhao, Shandong, China
| | - Shaoqing Sun
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Shandong, China
| | - Xingyao Hao
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Shandong, China
| | - Yong Wen
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Shandong, China.
| |
Collapse
|
35
|
Wang X, Zhang L, Dong B. Molecular mechanisms in MASLD/MASH-related HCC. Hepatology 2024:01515467-990000000-00739. [PMID: 38349726 PMCID: PMC11323288 DOI: 10.1097/hep.0000000000000786] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/16/2024] [Indexed: 03/23/2024]
Abstract
Liver cancer is the third leading cause of cancer-related deaths and ranks as the sixth most prevalent cancer type globally. NAFLD or metabolic dysfunction-associated steatotic liver disease, and its more severe manifestation, NASH or metabolic dysfunction-associated steatohepatitis (MASH), pose a significant global health concern, affecting approximately 20%-25% of the population. The increased prevalence of metabolic dysfunction-associated steatotic liver disease and MASH is parallel to the increasing rates of obesity-associated metabolic diseases, including type 2 diabetes, insulin resistance, and fatty liver diseases. MASH can progress to MASH-related HCC (MASH-HCC) in about 2% of cases each year, influenced by various factors such as genetic mutations, carcinogen exposure, immune microenvironment, and microbiome. MASH-HCC exhibits distinct molecular and immune characteristics compared to other causes of HCC and affects both men and women equally. The management of early to intermediate-stage MASH-HCC typically involves surgery and locoregional therapies, while advanced HCC is treated with systemic therapies, including anti-angiogenic therapies and immune checkpoint inhibitors. In this comprehensive review, we consolidate previous research findings while also providing the most current insights into the intricate molecular processes underlying MASH-HCC development. We delve into MASH-HCC-associated genetic variations and somatic mutations, disease progression and research models, multiomics analysis, immunological and microenvironmental impacts, and discuss targeted/combined therapies to overcome immune evasion and the biomarkers to recognize treatment responders. By furthering our comprehension of the molecular mechanisms underlying MASH-HCC, our goal is to catalyze the advancement of more potent treatment strategies, ultimately leading to enhanced patient outcomes.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Liang Zhang
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bingning Dong
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
36
|
Xu R, Liu X, Zhang Y, Wang K, Chen Z, Zheng J, Zhang T, Tong P, Qian Y, Yang W. Activating transcriptional coactivator with PDZ-binding motif by (R)-PFI-2 attenuates osteoclastogenesis and prevents ovariectomized-induced osteoporosis. Biochem Pharmacol 2024; 219:115964. [PMID: 38049011 DOI: 10.1016/j.bcp.2023.115964] [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: 08/26/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/06/2023]
Abstract
Excessive osteoclast activation is a leading cause of osteoporosis. Therefore, identifying molecular targets and relevant pharmaceuticals that inhibit osteoclastogenesis is of substantial clinical importance. Prior research has indicated that transcriptional coactivator with PDZ-binding motif (TAZ) impedes the process of osteoclastogenesis by engaging the nuclear factor (NF)-κB signaling pathway, thereby suggesting TAZ activation as a potential therapeutic approach to treat osteoporosis. (R)-PFI-2 is a novel selective inhibitor of SETD7 methyltransferase activity, which prevents the nuclear translocation of YAP, a homolog of TAZ. Therefore, we hypothesized that (R)-PFI-2 could be an effective therapeutic agent in the treatment of osteoporosis. To test this hypothesis and explore the underlying mechanism, we first examined the impact of (R)-PFI-2 on osteoclastogenesis in bone marrow macrophages (BMMs) in vitro. (R)-PFI-2 treatment inhibited TAZ phosphorylation induced by NF-κB, thereby enhancing its nuclear localization, protein expression, and activation in BMMs. Moreover, (R)-PFI-2-induced TAZ activation inhibited osteoclast formation in a dose-dependent manner, which involved inhibition of osteoclastogenesis through the TAZ and downstream NF-κB pathways. Furthermore, (R)-PFI-2 inhibited osteoclastogenesis and prevented ovariectomy-induced bone loss in vivo in a mouse model. Overall, our findings suggest that TAZ activation by (R)-PFI-2 inhibits osteoclastogenesis and prevents osteoporosis, indicating an effective strategy for treating osteoclast-induced osteoporosis.
Collapse
Affiliation(s)
- Rongjian Xu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China; Department of Orthopedics, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang Province 312000, China
| | - Xuewen Liu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China; Department of Orthopedics, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang Province 312000, China
| | - Yufeng Zhang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China; Department of Orthopedics, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang Province 312000, China
| | - Kelei Wang
- Department of Orthopedics, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang Province 312000, China
| | - Zhuolin Chen
- Department of Orthopedics, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang Province 312000, China
| | - Jiewen Zheng
- Department of Orthopedics, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang Province 312000, China
| | - Tan Zhang
- Department of Orthopedics, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang Province 312000, China
| | - Peijian Tong
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, China.
| | - Yu Qian
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China; Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, China.
| | - Wanlei Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
37
|
Benhammou JN, Qiao B, Ko A, Sinnett-Smith J, Pisegna JR, Rozengurt E. Lipophilic statins inhibit YAP coactivator transcriptional activity in HCC cells through Rho-mediated modulation of actin cytoskeleton. Am J Physiol Gastrointest Liver Physiol 2023; 325:G239-G250. [PMID: 37366601 PMCID: PMC10511177 DOI: 10.1152/ajpgi.00089.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of liver-related death. Lipophilic statins have been associated with a decrease in HCC incidence, raising the possibility of their use as chemoprevention agents. The Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) have emerged as an important pro-oncogenic mechanism in HCC. Statins modulate YAP/TAZ in other solid tumors, but few studies have assessed their mechanisms in HCC. We aimed to delineate how lipophilic statins regulate YAP protein localization by interrogating the mevalonate pathway in a stepwise manner using pharmacological and genetical approaches in HCC cells. Huh7 and Hep3B HCC cells were treated with the lipophilic statins cerivastatin and atorvastatin. YAP protein localization was determined using quantitative immunofluorescence (IF) imaging. The gene expression of CTGF and CYR61, known YAP/TEA-domain DNA-binding factor (TEAD)-regulated genes, was measured using quantitative real-time PCR. Rescue experiments were conducted using metabolites of the mevalonate pathway including mevalonic acid and geranylgeranyl pyrophosphate (GG-PP). The cellular cytoskeleton was assessed using F-actin IF staining. YAP protein was extruded from the nucleus to the cytoplasm with statin treatment. Consistently, CTGF and CYR61 mRNA expression significantly decreased with statins. Cytoskeletal structure was also compromised with statins. Gene expression, YAP protein localization, and cytoskeletal structure were all restored to baseline with exogenous GG-PP but not with other metabolites of the mevalonate pathway. Direct Rho GTPase inhibitor treatment mirrored the statin effects on YAP. YAP protein localization is regulated by lipophilic statins via Rho GTPases, causing cytoskeletal structural changes and is independent of cholesterol metabolites.NEW & NOTEWORTHY Statins are widely used for the treatment of cardiovascular diseases. Recently, their use has been associated with a decrease in the incidence of hepatocellular carcinoma (HCC); however, their mechanism(s) has remained elusive. In this study, we delineate the mechanism by which statins affect the Yes-associated protein (YAP), which has emerged as a key oncogenic pathway in HCC. We investigate each step of the mevalonate pathway and demonstrate that statins regulate YAP via Rho GTPases.
Collapse
Affiliation(s)
- Jihane N Benhammou
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, California, United States
- Division of Gastroenterology, Hepatology and Parental Nutrition, Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, United States
| | - Bo Qiao
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, California, United States
- Division of Gastroenterology, Hepatology and Parental Nutrition, Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
| | - Arthur Ko
- Center for Genetic Medicine Research, Childrens National Research Institute, Washington, District of Columbia, United States
| | - James Sinnett-Smith
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, California, United States
- Division of Gastroenterology, Hepatology and Parental Nutrition, Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
| | - Joseph R Pisegna
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, California, United States
- Division of Gastroenterology, Hepatology and Parental Nutrition, Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, United States
| | - Enrique Rozengurt
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, California, United States
- Division of Gastroenterology, Hepatology and Parental Nutrition, Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, United States
| |
Collapse
|
38
|
Adlat S, Vázquez Salgado AM, Lee M, Yin D, Wangensteen KJ. Emerging and potential use of CRISPR in human liver disease. Hepatology 2023:01515467-990000000-00538. [PMID: 37607734 PMCID: PMC10881897 DOI: 10.1097/hep.0000000000000578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/13/2023] [Indexed: 08/24/2023]
Abstract
CRISPR is a gene editing tool adapted from naturally occurring defense systems from bacteria. It is a technology that is revolutionizing the interrogation of gene functions in driving liver disease, especially through genetic screens and by facilitating animal knockout and knockin models. It is being used in models of liver disease to identify which genes are critical for liver pathology, especially in genetic liver disease, hepatitis, and in cancer initiation and progression. It holds tremendous promise in treating human diseases directly by editing DNA. It could disable gene function in the case of expression of a maladaptive protein, such as blocking transthyretin as a therapy for amyloidosis, or to correct gene defects, such as restoring the normal functions of liver enzymes fumarylacetoacetate hydrolase or alpha-1 antitrypsin. It is also being studied for treatment of hepatitis B infection. CRISPR is an exciting, evolving technology that is facilitating gene characterization and discovery in liver disease and holds the potential to treat liver diseases safely and permanently.
Collapse
Affiliation(s)
- Salah Adlat
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | | | |
Collapse
|