1
|
Wang Z, Li Y, Wang X, Zhang W, Chen Y, Lu X, Jin C, Tu L, Jiang T, Yang Y, Ma X, Zeng J, Wen Y, Efferth T. Precision Strike Strategy for Liver Diseases Trilogy with Xiao-Chai-Hu Decoction: A Meta-Analysis with Machine Learning. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156796. [PMID: 40347886 DOI: 10.1016/j.phymed.2025.156796] [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: 01/20/2025] [Revised: 03/30/2025] [Accepted: 04/20/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND AND PURPOSE The progression from hepatitis to liver fibrosis (LF) and ultimately to hepatic carcinoma (HCC) represents the advanced stages of various liver diseases. Currently, no universal treatment effectively addresses all three conditions. The Traditional Chinese Medicine formula Xiao-Chai-Hu decoction (XCHD) has shown promise in treating hepatitis, inhibiting LF, and serving as an adjunct therapy for HCC. This study evaluates the efficacy and optimal treatment durations of XCHD in managing these liver diseases using meta-analysis and machine learning techniques. METHODS Registered in the PROSPERO database (CRD42024534445), this meta-analysis systematically searched seven databases, including 54 studies with a total of 5,710 patients. Statistical analysis was performed using Stata 17.0. Five machine learning models-Random Forest (RF), XGBoost, Lasso, Multilayer Perceptron (MLP), and a stacking model combining these algorithms-were employed to analyze the data and predict the time-effect relationships. The optimal durations of XCHD treatment for the liver disease trilogy were subsequently projected. RESULTS XCHD significantly improved the primary outcome indicators for hepatitis, liver fibrosis, and HCC. Additionally, XCHD demonstrated a beneficial effect on liver dysfunction caused by these diseases. Machine learning predicted the optimal treatment durations of XCHD as 12 weeks for hepatitis, 20.31 weeks for liver fibrosis, and 12 weeks for HCC. CONCLUSION XCHD is effective in treating the liver disease trilogy, with optimal treatment durations of 12 weeks for hepatitis and HCC, and 20.31 weeks for liver fibrosis. These findings support the potential of XCHD in developing precise clinical strategies for managing liver diseases. This study innovatively integrates meta-analysis with machine learning to determine the optimal treatment durations, providing a novel approach for evidence-based precision medicine in Traditional Chinese Medicine.
Collapse
Affiliation(s)
- Zexin Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yubing Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaobao Wang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Chunmei Jin
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Lang Tu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tao Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiqin Yang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
2
|
Laman Trip DS, van Oostrum M, Memon D, Frommelt F, Baptista D, Panneerselvam K, Bradley G, Licata L, Hermjakob H, Orchard S, Trynka G, McDonagh EM, Fossati A, Aebersold R, Gstaiger M, Wollscheid B, Beltrao P. A tissue-specific atlas of protein-protein associations enables prioritization of candidate disease genes. Nat Biotechnol 2025:10.1038/s41587-025-02659-z. [PMID: 40316700 DOI: 10.1038/s41587-025-02659-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 03/28/2025] [Indexed: 05/04/2025]
Abstract
Despite progress in mapping protein-protein interactions, their tissue specificity is understudied. Here, given that protein coabundance is predictive of functional association, we compiled and analyzed protein abundance data of 7,811 proteomic samples from 11 human tissues to produce an atlas of tissue-specific protein associations. We find that this method recapitulates known protein complexes and the larger structural organization of the cell. Interactions of stable protein complexes are well preserved across tissues, while cell-type-specific cellular structures, such as synaptic components, are found to represent a substantial driver of differences between tissues. Over 25% of associations are tissue specific, of which <7% are because of differences in gene expression. We validate protein associations for the brain through cofractionation experiments in synaptosomes, curation of brain-derived pulldown data and AlphaFold2 modeling. We also construct a network of brain interactions for schizophrenia-related genes, indicating that our approach can functionally prioritize candidate disease genes in loci linked to brain disorders.
Collapse
Affiliation(s)
- Diederik S Laman Trip
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Marc van Oostrum
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Danish Memon
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Fabian Frommelt
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Delora Baptista
- Gulbenkian Institute for Molecular Medicine, Oeiras, Portugal
| | - Kalpana Panneerselvam
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Glyn Bradley
- Computational Biology, Functional Genomics, GSK, Stevenage, UK
| | - Luana Licata
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Henning Hermjakob
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Sandra Orchard
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Gosia Trynka
- Open Targets, Wellcome Genome Campus, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Ellen M McDonagh
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Andrea Fossati
- Science for Life Laboratory, Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Solna, Sweden
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Bernd Wollscheid
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
| | - Pedro Beltrao
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK.
- Open Targets, Wellcome Genome Campus, Cambridge, UK.
- Gulbenkian Institute for Molecular Medicine, Oeiras, Portugal.
| |
Collapse
|
3
|
Chen J, Yang Z, Cui Y, Zhao Z, Deng D, Fu Z, Zhang X. Increased expression of DNAJC7 promotes the progression of hepatocellular carcinoma by influencing the cell cycle and immune microenvironment. J Cancer Res Clin Oncol 2025; 151:154. [PMID: 40312488 PMCID: PMC12045834 DOI: 10.1007/s00432-025-06202-0] [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: 03/08/2025] [Accepted: 04/12/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the leading cause of cancer-related mortality worldwide owing to the lack of effective and early diagnostic tools and therapeutic approaches. DNAJC7, a member of the DnaJ heat shock family, is crucial in protein folding and stability; however, its specific functions and mechanisms in HCC remain unclear. OBJECTIVE This study aimed to explore the role of DNAJC7 in HCC progression and evaluate its potential clinical significance as a prognostic marker. METHODS Public databases (TCGA, ICGC, GEO, and TIMER) were used to assess DNAJC7 expression, correlations with clinical parameters, and related signaling pathways. Proliferation, migration, invasion, and cell cycle assays were performed to evaluate the function of DNAJC7 in HCC. Immune infiltration and associations with checkpoint proteins were analyzed using TIMER, and a Gene Set Enrichment Analysis (GSEA) was used to explore enriched pathways. RESULTS DNAJC7 expression was higher in HCC tissues than in adjacent normal tissues and was associated with advanced malignancy and poor prognosis, including a lower overall survival, progression-free survival, and disease-free survival. DNAJC7 knockdown resulted in reduced malignant behavior of HCC cells, leading to S-phase cell cycle arrest. Increased DNAJC7 expression was associated with immune cell infiltration and the presence of immunological checkpoint molecules, including CTLA4 and PD-1. GSEA highlighted the activation of key pathways, including WNT signaling and cell cycle regulation. CONCLUSION DNAJC7 regulates tumor cell proliferation, migration, invasion, and immune evasion by acting as an oncogene in HCC. It can serve as a diagnostic and prognostic biomarker and potential treatment target for HCC.
Collapse
Affiliation(s)
- Jiaxing Chen
- Department of Hepatobiliary Pancreatic Surgery, Henan Provincial People's Hospital, Zhengzhou City, 450003, Henan Province, China
| | - Zhizhao Yang
- Hepatobiliary Pancreatic Surgery Department of Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou City, 450003, Henan Province, China
| | - Yongqiang Cui
- Hepatobiliary Pancreatic Surgery Department of Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou City, 450003, Henan Province, China
| | - Zhilei Zhao
- Hepatobiliary Pancreatic Surgery Department of Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou City, 450003, Henan Province, China
| | - Dongfeng Deng
- Department of Hepatobiliary Pancreatic Surgery, Henan Provincial People's Hospital, Zhengzhou City, 450003, Henan Province, China
| | - Zhihao Fu
- Department of Hepatobiliary Pancreatic Surgery, Henan Provincial People's Hospital, Zhengzhou City, 450003, Henan Province, China
| | - Xiao Zhang
- Department of Hepatobiliary Pancreatic Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital and Henan University People's Hospital, No. 7 Weiwu Road, Jinshui District, Zhengzhou City, 450003, Henan Province, China.
| |
Collapse
|
4
|
Chen QF, Chen S, Zhao M. The Synergistic Mechanisms and Prospects of Transarterial Chemoembolization Combined with Immunotherapy for Hepatocellular Carcinoma. J Hepatocell Carcinoma 2025; 12:841-854. [PMID: 40322278 PMCID: PMC12050036 DOI: 10.2147/jhc.s514881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
Hepatocellular carcinoma (HCC) represents a highly aggressive form of liver neoplasm that presents various therapeutic obstacles. Recently, the synergistic use of transarterial chemoembolization (TACE) in conjunction with immunotherapy has attracted considerable interest within the medical community. This review aims to explore the synergistic mechanisms between TACE and immunotherapy, analyze the current research evidence, and discuss their potential applications in the treatment of HCC. By examining how TACE can enhance the efficacy of immunotherapy, we seek to provide direction for future research and emphasize the importance of personalized treatment strategies in managing HCC.
Collapse
Affiliation(s)
- Qi-Feng Chen
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, People’s Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong, People’s Republic of China
| | - Song Chen
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, People’s Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong, People’s Republic of China
| | - Ming Zhao
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, People’s Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
5
|
Kato M, Nishino J, Nagai M, Rokutan H, Narushima D, Ono H, Hasegawa T, Imoto S, Matsui S, Tsunoda T, Shibata T. Comprehensive analysis of prognosis markers with molecular features derived from pan-cancer whole-genome sequences. Hum Genomics 2025; 19:39. [PMID: 40221813 PMCID: PMC11993945 DOI: 10.1186/s40246-025-00744-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Cancer prognosis markers are useful for treatment decisions; however, the omics-level landscape is not well understood across multiple cancer types. Pan-Cancer Analysis of Whole Genomes (PCAWG) provides unprecedented access to various types of molecular data, ranging from typical DNA mutations and RNA expressions to more deeply analyzed or whole-genomic features, such as HLA haplotypes and structural variations. We analyzed the PCAWG data of 13 cancer types from 1,514 patients to identify prognosis markers belonging to 17 molecular features in the survival analysis based on the Cox and Lasso regression methods. We found that germline features including HLA haplotypes, neoantigens, and the number of structural variations were associated with overall survival; however, mutational signatures were not. Measuring a few markers provided a sufficient prognostic performance evaluated by c-index for each cancer type. DNA markers demonstrated better or comparable prognostic performance compared to RNA markers in some cancer types. "Universal" markers strongly associated with overall survival across cancer types were not identified. These findings will give insights into the clinical implementation of prognosis markers.
Collapse
Affiliation(s)
- Mamoru Kato
- Division of Bioinformatics, Research Institute, National Cancer Center Japan, Tokyo, Japan.
- CREST, JST, Tokyo, Japan.
| | - Jo Nishino
- Division of Bioinformatics, Research Institute, National Cancer Center Japan, Tokyo, Japan
- CREST, JST, Tokyo, Japan
| | - Momoko Nagai
- Division of Bioinformatics, Research Institute, National Cancer Center Japan, Tokyo, Japan
- CREST, JST, Tokyo, Japan
| | - Hirofumi Rokutan
- Division of Cancer Genomics, Research Institute, National Cancer Center Japan, Tokyo, Japan
| | - Daichi Narushima
- Division of Bioinformatics, Research Institute, National Cancer Center Japan, Tokyo, Japan
| | - Hanako Ono
- Division of Bioinformatics, Research Institute, National Cancer Center Japan, Tokyo, Japan
| | - Takanori Hasegawa
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shigeyuki Matsui
- CREST, JST, Tokyo, Japan
- Department of Biostatistics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tatsuhiko Tsunoda
- CREST, JST, Tokyo, Japan
- Laboratory for Medical Science Mathematics, Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tatsuhiro Shibata
- Division of Cancer Genomics, Research Institute, National Cancer Center Japan, Tokyo, Japan
- Laboratory of Molecular Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Wang M, Li Y, Su J, Dong X, Liu A, Yang Y, Tang X, Chen R, Li Q, Wang H, Xiao H. Endogenous protein S100A14 stabilizes glutaminase to render hepatocellular carcinoma resistant to sorafenib. J Transl Med 2025; 23:435. [PMID: 40217256 PMCID: PMC11992768 DOI: 10.1186/s12967-025-06333-5] [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: 12/10/2024] [Accepted: 03/01/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Many cases of advanced hepatocellular carcinoma (HCC) are resistant to the widely used drug sorafenib, which worsens prognosis. While many studies have explored how acquired resistance emerges during drug exposure, the mechanism underlying primary resistance before treatment still remain elusive. METHODS Single-cell lineage tracing and RNA sequencing were performed to identify primary sorafenib-resistant lineages in HCC. Differential gene expression analysis was employed to identify the biomarkers of drug-resistant lineage cells. Cell viability and colony formation assays were adopted to assess the involvement of S100A14 on sorafenib resistance. Co-immunoprecipitation (CO-IP) and mass spectrometry analysis were conducted to uncover the downstream targets and regulatory mechanisms of S100A14 in primary resistance to sorafenib. In vivo mouse xenograft experiments were carried out to assess the effect of S100A14 or its interacting protein glutaminase (GLS) on primary resistance to sorafenib in HCC. RESULTS Single-cell lineage tracing identified a cluster of sorafenib primary resistant cells, and S100A14, a Ca2+-binding protein, was determined to be a critical biomarker for primary resistance to sorafenib. Knockdown of S100A14 significantly increases sorafenib treatment sensitivity in HCC cells. Mechanistically, S100A14 binds to GLS and blocks its phosphorylation at residues Y308 and S314, which in turn inhibits its ubiquitination and subsequent degradation. By stabilizing GLS, S100A14 reduces oxidative stress in HCC cells, thereby antagonizing sorafenib-induced apoptosis. Inhibiting S100A14 or GLS significantly improved sorafenib efficacy against xenograft tumors in vivo. CONCLUSIONS Our results demonstrate that S100A14 plays a pivotal role in promoting primary resistance to sorafenib by stabilizing GLS to reduce oxidative stress, and acts as a potential therapeutic target to enhance the efficacy of sorafenib in HCC patients.
Collapse
Affiliation(s)
- Menghui Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yueheng Li
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Junhui Su
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Xinjue Dong
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Ao Liu
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Yuqi Yang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Xinyi Tang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Ruijie Chen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - QingQuan Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China.
| | - Hongshan Wang
- Department of General Surgery, GI Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200030, P. R. China.
- Department of General Surgery, Shanghai Baoshan District Wusong Central Hospital (Zhongshan Hospital Wusong Branch Fudan University), Shanghai, P. R. China.
- Baoshan Cancer Center, Baoshan District, Shanghai, P. R. China.
| | - Hong Xiao
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, P. R. China.
| |
Collapse
|
7
|
Wang YY, Yang WX, Cai JY, Wang FF, You CG. Comprehensive molecular characteristics of hepatocellular carcinoma based on multi-omics analysis. BMC Cancer 2025; 25:573. [PMID: 40159482 PMCID: PMC11956240 DOI: 10.1186/s12885-025-13952-0] [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: 10/17/2024] [Accepted: 03/17/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND The high heterogeneity of hepatocellular carcinoma (HCC) poses challenges for precision treatment strategies. This study aims to use multi-omics methodologies to better understand its pathogenesis and discover biomarkers. METHODS Quantitative proteomics was used to investigate hepatocellular carcinoma tissues (HCT) and their corresponding adjacent non-tumor tissues (DNT), obtained from six HCC patients. Untargeted metabolomics was applied to analyze the metabolic profiles of HCT and DNT of ten HCC patients. Statistical analyses, such as the Student's t-test, were performed to identify differentially expressed proteins (DEPs) and metabolites (DEMs) between the two groups. The functions and metabolic pathways involving DEPs and DEMs were annotated and enriched using the gene ontology (GO) and kyoto encyclopedia of genes and genomes (KEGG) databases. Bioinformatics methods were then utilized to analyze consistency between proteomics and metabolomics results, leading to identification of potential biomarkers along with key altered pathways associated with HCC. RESULTS This study identified 1556 DEPs between HCT and DNT samples. These DEPs were primarily enriched in crucial biological pathways such as amino acid degradation, fatty acid metabolism, and DNA replication. Subsequently, the analysis of metabolomics identified 500 DEMs that mainly participated in glycerophospholipid metabolism, the phospholipase D signaling pathway, and choline metabolism related to cancer. Integrated analysis of proteomics and metabolomics data unveiled significant dysfunctions in bile secretion, multiple amino acid and fatty acid metabolic pathways among HCC patients. Further investigation revealed that five proteins (PTP4A3, B4GALT5, GAB1, ME2, and PKM) along with seven metabolites (PI(6 keto-PGF1alpha/16:0), 13, 16, 19-docosatrienoic acid, PA(18:2(9Z, 12Z)/20:1(11Z)), Citric Acid, PG(20:3(6, 8, 11)-OH(5)/18:2(9Z, 12Z)), Spermidine, and N2-Acetylornithine) exhibited excellent diagnostic efficiency for HCC and could serve as its potential biomarkers. CONCLUSION Our integrated proteome and metabolome analysis revealed 10 key HCC-related pathways and proposed 12 potential biomarkers, which may enhance our understanding of HCC pathophysiology and be helpful in facilitating early diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Ying-Ying Wang
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, 730030, China
| | - Wan-Xia Yang
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, 730030, China
| | - Jiang-Ying Cai
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, 730030, China
| | - Fang-Fang Wang
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, 730030, China
| | - Chong-Ge You
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, 730030, China.
| |
Collapse
|
8
|
Cannet F, Sequera C, Veloso PM, El Kaoutari A, Methia M, Richelme S, Kaya M, Cherni A, Dupont M, Borg JP, Morel C, Boursier Y, Maina F. Tracing specificity of immune landscape remodeling associated with distinct anticancer treatments. iScience 2025; 28:112071. [PMID: 40124507 PMCID: PMC11930375 DOI: 10.1016/j.isci.2025.112071] [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: 03/03/2024] [Revised: 07/18/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Immune cells within the tumor microenvironment impact cancer progression, resistance, response to treatments. Despite remarkable outcomes for some cancer patients, immunotherapies remain unsatisfactory for others. Here, we designed an experimental setting using the Alb-R26 Met "inside-out" mouse model, faithfully recapitulating molecular features of liver cancer patients, to explore the effects of distinct anticancer targeted therapies on the tumor immune landscape. Using two treatments in clinical trials for different cancer types, Decitabine and MEK+BCL-XL blockage, we show their capability to trigger tumor regression in Alb-R26 Met mice and to superimpose distinct profiles of immune cell types and immune-checkpoints, impacting immunotherapy response. A machine learning approach processing tumor imaging and immune profile data identified a putative signature predicting tumor treatment response in mice and patients. Outcomes exemplify how the tumor immune microenvironment is differentially reshaped by distinct anticancer agents and highlight the importance of measuring its modulation during treatment to optimize oncotherapy and immunotherapy combinations.
Collapse
Affiliation(s)
- Floriane Cannet
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Célia Sequera
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Paula Michea Veloso
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Abdessamad El Kaoutari
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Melissa Methia
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Sylvie Richelme
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Muge Kaya
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Afef Cherni
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | - Mathieu Dupont
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | - Jean-Paul Borg
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Institut Universitaire de France, Paris, France
| | - Christian Morel
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | | | - Flavio Maina
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| |
Collapse
|
9
|
Hernandez R, Garcia-Rodriguez NS, Arriaga MA, Perez R, Bala AA, Leandro AC, Diego VP, Almeida M, Parsons JG, Manusov EG, Galan JA. The hepatocellular model of fatty liver disease: from current imaging diagnostics to innovative proteomics technologies. Front Med (Lausanne) 2025; 12:1513598. [PMID: 40109726 PMCID: PMC11919916 DOI: 10.3389/fmed.2025.1513598] [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: 10/18/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is a prevalent chronic liver condition characterized by lipid accumulation and inflammation, often progressing to severe liver damage. We aim to review the pathophysiology, diagnostics, and clinical care of MASLD, and review highlights of advances in proteomic technologies. Recent advances in proteomics technologies have improved the identification of novel biomarkers and therapeutic targets, offering insight into the molecular mechanisms underlying MASLD progression. We focus on the application of mass spectrometry-based proteomics including single cell proteomics, proteogenomics, extracellular vesicle (EV-omics), and exposomics for biomarker discovery, emphasizing the potential of blood-based panels for noninvasive diagnosis and personalized medicine. Future research directions are presented to develop targeted therapies and improve clinical outcomes for MASLD patients.
Collapse
Affiliation(s)
- Renee Hernandez
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Natasha S Garcia-Rodriguez
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Marco A Arriaga
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Ricardo Perez
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Auwal A Bala
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Ana C Leandro
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Vince P Diego
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Marcio Almeida
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Jason G Parsons
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Eron G Manusov
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Jacob A Galan
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| |
Collapse
|
10
|
Dhanasekaran R, Suzuki H, Lemaitre L, Kubota N, Hoshida Y. Molecular and immune landscape of hepatocellular carcinoma to guide therapeutic decision-making. Hepatology 2025; 81:1038-1057. [PMID: 37300379 PMCID: PMC10713867 DOI: 10.1097/hep.0000000000000513] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023]
Abstract
Liver cancer, primarily HCC, exhibits highly heterogeneous histological and molecular aberrations across tumors and within individual tumor nodules. Such intertumor and intratumor heterogeneities may lead to diversity in the natural history of disease progression and various clinical disparities across the patients. Recently developed multimodality, single-cell, and spatial omics profiling technologies have enabled interrogation of the intertumor/intratumor heterogeneity in the cancer cells and the tumor immune microenvironment. These features may influence the natural history and efficacy of emerging therapies targeting novel molecular and immune pathways, some of which had been deemed undruggable. Thus, comprehensive characterization of the heterogeneities at various levels may facilitate the discovery of biomarkers that enable personalized and rational treatment decisions, and optimize treatment efficacy while minimizing the risk of adverse effects. Such companion biomarkers will also refine HCC treatment algorithms across disease stages for cost-effective patient management by optimizing the allocation of limited medical resources. Despite this promise, the complexity of the intertumor/intratumor heterogeneity and ever-expanding inventory of therapeutic agents and regimens have made clinical evaluation and translation of biomarkers increasingly challenging. To address this issue, novel clinical trial designs have been proposed and incorporated into recent studies. In this review, we discuss the latest findings in the molecular and immune landscape of HCC for their potential and utility as biomarkers, the framework of evaluation and clinical application of predictive/prognostic biomarkers, and ongoing biomarker-guided therapeutic clinical trials. These new developments may revolutionize patient care and substantially impact the still dismal HCC mortality.
Collapse
Affiliation(s)
| | - Hiroyuki Suzuki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka
| | - Lea Lemaitre
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California
| | - Naoto Kubota
- Liver Tumor Translational Research Program, Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yujin Hoshida
- Liver Tumor Translational Research Program, Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
11
|
Cheng Q, Han X, Xie H, Liao YL, Wang F, Cui XY, Jiang , Zhang CW. PAXIP1 is regulated by NRF1 and is a prognosis‑related biomarker in hepatocellular carcinoma. Biomed Rep 2025; 22:38. [PMID: 39781045 PMCID: PMC11704871 DOI: 10.3892/br.2024.1916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is characterized by a poor prognosis globally. PAX-interacting protein 1 (PAXIP1) serves a key role in the development of numerous human cancer types. Nevertheless, its specific involvement in HCC remains poorly understood. Public repository systems (Integrative Molecular Database of HCC, Gene Expression Omnibus, The Cancer Genome Atlas, University of Alabama at Birmingham Cancer Data Analysis Portal, Tumor Immune Estimation Resource and Human Protein Atlas) were utilized to explore PAXIP1 expression in HCC and evaluate the prognostic value of PAXIP1 in patients with HCC. PAXIP1 expression was investigated, and a notable relationship between PAXIP1 expression and various cancer types was found through analysis of The Cancer Genome Atlas data. More specifically, patients with HCC and lower PAXIP1 levels had improved survival rates. Furthermore, using LinkedOmics, the co-expression network of PAXIP1 in HCC was determined. Colocalization analysis of PAXIP1 using chromatin immunoprecipitation-sequencing data suggested that PAXIP1 might act as a cofactor for MYB proto-oncogene like 2 or FOXO1 in HCC. In addition, by predicting and analyzing the potential transcription factors related to PAXIP1, nuclear respiratory factor 1 was identified as a factor upstream of PAXIP1 in HCC. Notably, PAXIP1 expression exhibited a positive association with the infiltration of CD4+ and CD8+ T cells, macrophages, neutrophils and myeloid dendritic cells. Furthermore, PAXIP1 expression was associated with a range of immune markers such as programmed cell death protein 1, programmed death-ligand 1 and cytotoxic T-lymphocyte associated protein 4 in HCC. The findings of the present study highlighted the prognostic relevance of PAXIP1 and its function in modulating immune cell recruitment in HCC.
Collapse
Affiliation(s)
- Qian Cheng
- Department of Pathogen Biology, Microbiology Division, Key Laboratory of Pathogen of Jiangsu Province Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Xiao Han
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Hao Xie
- School of Life Science and Technology, Southeast University, Nanjing, Jiangsu 210018, P.R. China
| | - Yan-Lin Liao
- MEDx (Suzhou) Translation Medicine Co., Ltd., Suzhou, Jiangsu 215000, P.R. China
| | - Fei Wang
- Wuxi Mental Health Center/Wuxi Central Rehabilitation Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Xiao-Ying Cui
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Chao Jiang
- Department of Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Cheng-Wan Zhang
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| |
Collapse
|
12
|
Suo Y, Song Y, Wang Y, Liu Q, Rodriguez H, Zhou H. Advancements in proteogenomics for preclinical targeted cancer therapy research. BIOPHYSICS REPORTS 2025; 11:56-76. [PMID: 40070661 PMCID: PMC11891078 DOI: 10.52601/bpr.2024.240053] [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: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 03/14/2025] Open
Abstract
Advancements in molecular characterization technologies have accelerated targeted cancer therapy research at unprecedented resolution and dimensionality. Integrating comprehensive multi-omic molecular profiling of a tumor, proteogenomics, marks a transformative milestone for preclinical cancer research. In this paper, we initially provided an overview of proteogenomics in cancer research, spanning genomics, transcriptomics, and proteomics. Subsequently, the applications were introduced and examined from different perspectives, including but not limited to genetic alterations, molecular quantifications, single-cell patterns, different post-translational modification levels, subtype signatures, and immune landscape. We also paid attention to the combined multi-omics data analysis and pan-cancer analysis. This paper highlights the crucial role of proteogenomics in preclinical targeted cancer therapy research, including but not limited to elucidating the mechanisms of tumorigenesis, discovering effective therapeutic targets and promising biomarkers, and developing subtype-specific therapies.
Collapse
Affiliation(s)
- Yuying Suo
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanli Song
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuqiu Wang
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Qian Liu
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Rockville, MD 20850, USA
| | - Hu Zhou
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
13
|
Prakash A, Collins A, Vilmovsky L, Fexova S, Jones AR, Vizcaino JA. Integrated View of Baseline Protein Expression in Human Tissues Using Public Data Independent Acquisition Data Sets. J Proteome Res 2025; 24:685-695. [PMID: 39764611 PMCID: PMC11811993 DOI: 10.1021/acs.jproteome.4c00788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/18/2024] [Accepted: 12/19/2024] [Indexed: 02/08/2025]
Abstract
The PRIDE database is the largest public data repository of mass spectrometry-based proteomics data and currently stores more than 40,000 data sets covering a wide range of organisms, experimental techniques, and biological conditions. During the past few years, PRIDE has seen a significant increase in the amount of submitted data-independent acquisition (DIA) proteomics data sets. This provides an excellent opportunity for large-scale data reanalysis and reuse. We have reanalyzed 15 public label-free DIA data sets across various healthy human tissues to provide a state-of-the-art view of the human proteome in baseline conditions (without any perturbations). We computed baseline protein abundances and compared them across various tissues, samples, and data sets. Our second aim was to compare protein abundances obtained here from the results of previous analyses using human baseline data-dependent acquisition (DDA) data sets. We observed a good correlation across some tissues, especially in the liver and colon, but weak correlations were found in others, such as the lung and pancreas. The reanalyzed results including protein abundance values and curated metadata are made available to view and download from the resource Expression Atlas.
Collapse
Affiliation(s)
- Ananth Prakash
- European
Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, U.K.
| | - Andrew Collins
- Institute
of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, U.K.
| | - Liora Vilmovsky
- European
Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, U.K.
| | - Silvie Fexova
- European
Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, U.K.
| | - Andrew R. Jones
- Institute
of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, U.K.
| | - Juan Antonio Vizcaino
- European
Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, U.K.
| |
Collapse
|
14
|
Yan S, Fu P, Zhu Y, Li H, Shan R, Gong B. Whole transcriptome and proteome analyses identify ncRNAs and mRNAs to predict competing endogenous RNA networks in hepatitis B virus-induced hepatocellular carcinoma. Microb Pathog 2025; 199:107248. [PMID: 39710348 DOI: 10.1016/j.micpath.2024.107248] [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: 09/16/2024] [Revised: 12/11/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
The presence of the Hepatitis B virus (HBV) is considered as a valuable risk factor of hepatocellular carcinoma (HCC). To more deeply comprehend the molecular mechanism and transcriptome of HBV-induced HCC, we utilized tandem mass tagging (TMT)-based quantitative proteomics analysis and whole-transcriptome sequencing to analyze three sets of matched HepG2 hepatoma cells and HBV-positive HepAD38 cells. The differentially expressed (DE) proteins (1596), mRNAs (5263), miRNAs (581), lncRNAs (2672) and circRNAs (222) were subjected to differential expression and enrichment analyses in order to thoroughly assess the gene-regulatory circuits of HBV-induced HCC. Subsequently, the amounts of 321 DEproteins-DEmRNAs with common alterations were confirmed. According to functional pathway analysis, the DEproteins-DEmRNAs were primarily linked to signaling pathways, amino acid metabolism, and cellular function. Furthermore, the viability and significance of the ceRNA regulatory networks, LOC105377730/miR-4726-5p/FHL2 and hsa_circ_0001098/miR-2110/IGF2BP1, were randomly chosen and confirmed. Our work provides a valuable asset in terms of understanding regulatory activities at the RNA level, and might reveal fresh information about the fundamental mechanism and potential therapeutic targets of HBV-induced HCC.
Collapse
Affiliation(s)
- Shaoying Yan
- Department of Clinical Laboratory, Medical Center of Burn Plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Diagnosis of Infectious Diseases, Nanchang, Jiangxi, China
| | - Peng Fu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yali Zhu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Huiming Li
- Department of Clinical Laboratory, Medical Center of Burn Plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Renfeng Shan
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Binbin Gong
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
15
|
Liu Y, Fan Y, Gong R, Qiu M, Wei X, Lin Q, Zhou Z, Cao J, Jiang Y, Chen P, Chen B, Yang X, Wei Y, Zhang R, Wen Q, Yu H. Novel genetic variants in the NLRP3 inflammasome-related PANX1 and APP genes predict survival of patients with hepatitis B virus-related hepatocellular carcinoma. Clin Transl Oncol 2025; 27:630-641. [PMID: 39090420 PMCID: PMC11782428 DOI: 10.1007/s12094-024-03634-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND The nod-like receptor protein 3 (NLRP3) is one of the most characterized inflammasomes involved in the pathogenesis of several cancers, including hepatocellular carcinoma (HCC). However, the effects of genetic variants in the NLRP3 inflammasome-related genes on survival of hepatitis B virus (HBV)-related HCC patients are unclear. METHODS We performed multivariable Cox proportional hazards regression analysis to evaluate associations between 299 single-nucleotide polymorphisms (SNPs) in 16 NLRP3 inflammasome-related genes and overall survival (OS) of 866 patients with HBV-related HCC. We further performed expression quantitative trait loci (eQTL) analysis using the data from the GTEx project and 1000 Genomes projects, and performed differential expression analysis using the TCGA dataset to explore possible molecular mechanisms underlying the observed associations. RESULTS We found that two functional SNPs (PANX1 rs3020013 A > G and APP rs9976425 C > T) were significantly associated with HBV-related HCC OS with the adjusted hazard ratio (HR) of 0.83 [95% confidence interval (CI) = 0.73-0.95, P = 0.008], and 1.26 (95% CI = 1.02-1.55, P = 0.033), respectively. Moreover, the eQTL analysis revealed that the rs3020013 G allele was correlated with decreased mRNA expression levels of PANX1 in both normal liver tissues (P = 0.044) and whole blood (P < 0.001) in the GTEx dataset, and PANX1 mRNA expression levels were significantly higher in HCC samples and associated with a poorer survival of HCC patients. However, we did not observe such correlations for APP rs9976425. CONCLUSIONS These results indicated that SNPs in the NLRP3 inflammasome-related genes may serve as potential biomarkers for HBV-related HCC survival, once replicated by additional larger studies.
Collapse
Affiliation(s)
- Yingchun Liu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
- Key Cultivated Laboratory of Cancer Molecular Medicine of Guangxi Health Commission, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
| | - Yuman Fan
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, China
| | - Rongbin Gong
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, China
| | - Moqin Qiu
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
| | - Xiaoxia Wei
- Department of Clinical Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
| | - Qiuling Lin
- Department of Clinical Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
| | - Zihan Zhou
- Department of Cancer Prevention and Control, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
| | - Ji Cao
- Department of Cancer Prevention and Control, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
| | - Yanji Jiang
- Department of Scientific Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
| | - Peiqin Chen
- Editorial Department of Chinese Journal of Oncology Prevention and Treatment, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
| | - Bowen Chen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
| | - Xiaobing Yang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, China
| | - Yuying Wei
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China
| | - RuoXin Zhang
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Qiuping Wen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China.
- Key Cultivated Laboratory of Cancer Molecular Medicine of Guangxi Health Commission, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China.
| | - Hongping Yu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China.
- Key Cultivated Laboratory of Cancer Molecular Medicine of Guangxi Health Commission, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, 22 Shuangyong Road, Nanning, Guangxi, China.
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 22 Shuangyong Road, Guangxi, China.
| |
Collapse
|
16
|
Zhou T, Han X, Xiao C, Lei X, Lan X, Wei X, Liang Y, Wu H. Diagnostic accuracy of preoperative MRI in assessing macrotrabecular-massive subtype of hepatocellular carcinoma: a systematic review and meta-analysis. Eur Radiol 2025:10.1007/s00330-024-11344-9. [PMID: 39836200 DOI: 10.1007/s00330-024-11344-9] [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: 07/18/2024] [Revised: 10/23/2024] [Accepted: 12/08/2024] [Indexed: 01/22/2025]
Abstract
OBJECTIVES To determine the value of preoperative magnetic resonance imaging (MRI) in predicting macrotrabecular-massive hepatocellular carcinoma (MTM-HCC). MATERIALS AND METHODS A search was conducted on PubMed, Web of Science, Cochrane Library databases, and Embase for studies evaluating the performance of MRI in assessing MTM-HCC. The quality assessment of diagnostic studies (QUADAS-2) tool was used to assess the risk of bias. Diagnostic accuracy measures, including sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), and diagnostic odds ratio (DOR), were pooled. Summary receiver operating characteristic (SROC) curves with the area under the curve (AUC) were generated. Meta-regression analysis was performed to explore potential sources of heterogeneity. RESULTS A total of ten eligible studies including 2074 lesions in 2053 patients were analyzed. The pooled sensitivity, specificity, PLR, NLR, DOR, and AUC were 0.65 (0.52, 0.76), 0.88 (0.80, 0.94), 5.6 (3.70, 8.60), 0.40 (0.30, 0.53), 14 (10, 20), and 0.84 (0.81, 0.87), respectively. High heterogeneity was observed (I2 was 78.61% and 90.95% for sensitivity and specificity, respectively) along with a threshold effect (Spearman's correlation coefficient = 0.927, p < 0.001). Meta-regression analysis demonstrated that the MRI method (radiomics or non-radiomics) affected the heterogeneity. CONCLUSION MRI has diagnostic value for MTM-HCC due to its higher specificity and moderate sensitivity, but its clinical application remains suboptimal due to significant heterogeneity. Thus, further prospective studies with large sample sizes are needed to confirm these results. KEY POINTS Question What is the value of MRI for preoperatively predicting MTM-HCC? Findings Meta-regression analyses revealed that the MRI method (radiomics or non-radiomics) is a significant factor contributing to heterogeneity. Clinical relevance This study demonstrates the high diagnostic accuracy of MRI for early detection of MTM-HCC, which can assist in guiding individualized management.
Collapse
Affiliation(s)
- Tingwen Zhou
- Guangdong Medical University, Zhanjiang, China
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaorui Han
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chuyin Xiao
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaoxiao Lei
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xinxin Lan
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xinhua Wei
- Guangdong Medical University, Zhanjiang, China
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yingying Liang
- Guangdong Medical University, Zhanjiang, China
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Hongzhen Wu
- Guangdong Medical University, Zhanjiang, China.
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
| |
Collapse
|
17
|
Li F, Hu B, Zhang L, Liu Y, Wang J, Wu C, Wu S, Zhang Y, Yang X, Lu H. Phosphoproteomics profiling of sorafenib-resistant hepatocellular carcinoma patient-derived xenografts reveals potential therapeutic strategies. iScience 2025; 28:111657. [PMID: 39886465 PMCID: PMC11780156 DOI: 10.1016/j.isci.2024.111657] [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: 02/25/2024] [Revised: 07/15/2024] [Accepted: 12/18/2024] [Indexed: 02/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer with poor prognosis. Sorafenib, a first-line treatment for advanced HCC, has shown limited clinical benefits due to the onset of drug resistance. Thus, it is imperative to comprehend the mechanisms underlying sorafenib resistance and explore strategies to overcome or delay it. Here, we established HCC patient-derived xenograft (PDX) models with acquired resistance to sorafenib and performed comprehensive proteomic and phosphoproteomic analyses on these models. The active cell cycle pathway along with the active cyclin-dependent kinase CDK1 and DNA-dependent protein kinase PRKDC was identified through KEGG pathway enrichment and kinase substrate enrichment analyses. Upon investigating the potential of combining sorafenib with putative kinase inhibitors, we found that the combination displays synergistic anti-proliferative effects in the sorafenib-resistant liver cancer cell line, thus providing a proof of concept for phosphoproteomic-guided design of precision medicine.
Collapse
Affiliation(s)
- Feng Li
- Liver Cancer Institute and Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Bo Hu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Lei Zhang
- Liver Cancer Institute and Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yang Liu
- Liver Cancer Institute and Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Jun Wang
- Liver Cancer Institute and Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Changqing Wu
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Suiyi Wu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Ying Zhang
- Department of Chemistry and Key Laboratory of Glycoconjugates Research Ministry of Public Health, Fudan University, Shanghai, P.R. China
| | - Xinrong Yang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Haojie Lu
- Liver Cancer Institute and Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
- Department of Chemistry and Key Laboratory of Glycoconjugates Research Ministry of Public Health, Fudan University, Shanghai, P.R. China
| |
Collapse
|
18
|
Hermán-Sánchez N, Del Rio-Moreno M, Ciria R, Sánchez-Frias ME, Fernández-Barrena MG, Uriarte I, Chicano-Galvez E, Ortea I, Peralbo-Molina Á, Briceño J, Avila MA, Rodríguez-Perálvarez M, Luque RM, López-Cánovas JL, Gahete MD. Quantitative proteomic analysis unveils a critical role of VARS1 in hepatocellular carcinoma aggressiveness through the modulation of MAGI1 expression. Mol Cancer 2025; 24:15. [PMID: 39810176 PMCID: PMC11731432 DOI: 10.1186/s12943-024-02206-5] [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: 10/31/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) genetic/transcriptomic signatures have been widely described. However, its proteomic characterization is incomplete. We performed non-targeted quantitative proteomics of HCC samples and explored its clinical, functional, and molecular consequences. METHODS Non-targeted quantitative proteomics were performed on cytosolic and nuclear fractions of liver samples [HCC vs. non-tumour adjacent tissue (NTAT), n = 42 patients]. Changes were confirmed in 7 in silico HCC cohorts. Functional and molecular implications were evaluated on HCC-derived cell lines after silencing/overexpressing VARS1 and/or MAGI1. VARS1-overexpressing Hep3B cells were used for in vivo studies [Extreme Limiting Dilution Assay (ELDA) and orthotopic tumour formation]. Quantitative proteomics were performed on VARS1-overexpressing HCC cell lines. RESULTS Quantitative proteomics revealed the dysregulation of the cytosolic and nuclear proteomes in HCC, and defined two proteomic HCC subgroups, the most aggressive associated to the dysregulation of the aminoacyl-tRNA synthetases (ARSs). ARSs dysregulation was corroborated in in silico HCC cohorts and associated to poor prognosis. Patients with ARSs upregulation had genomic/transcriptomic characteristics of the proliferative HCC. Valine tRNA-aminoacyl synthetase (VARS1) was the ARSs most consistently overexpressed and associated to aggressiveness. VARS1 modulation (silencing/overexpression) altered tumour establishment-associated parameters in vitro and/or in vivo. Quantitative proteomics on cells overexpressing VARS1 and rescue experiments identified the downregulation of MAGI1, a tumour suppressor in HCC, as a mediator of VARS1 function. CONCLUSIONS Quantitative proteomics defines two prognosis-related proteomic HCC subgroups. ARSs machinery is dysregulated in the aggressive subgroup, bearing potential as prognostic biomarkers. VARS1 promotes aggressiveness through the modulation of MAGI1, representing a novel targetable vulnerability in HCC.
Collapse
Affiliation(s)
- Natalia Hermán-Sánchez
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Mercedes Del Rio-Moreno
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Rubén Ciria
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
- Unit of Hepatobiliary Surgery and Liver Transplantation, Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Marina E Sánchez-Frias
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
- Anatomical Pathology Department, Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Maite G Fernández-Barrena
- Hepatology Laboratory, Solid Tumors Program, CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), CIMA, University of Navarra, Instituto de Salud Carlos III, Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain
| | - Iker Uriarte
- Hepatology Laboratory, Solid Tumors Program, CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), CIMA, University of Navarra, Instituto de Salud Carlos III, Pamplona, Spain
| | - Eduardo Chicano-Galvez
- IMIBIC Mass Spectrometry and Molecular Imaging Unit (IMSMI), Reina Sofía University Hospital, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba (UCO), Cordoba, 14004, Spain
| | - Ignacio Ortea
- Centro de Investigación en Nanomateriales y Nanotecnología (CINN-CSIC), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, 33011, Spain
| | - Ángela Peralbo-Molina
- IMIBIC Mass Spectrometry and Molecular Imaging Unit (IMSMI), Reina Sofía University Hospital, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba (UCO), Cordoba, 14004, Spain
| | - Javier Briceño
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
- Unit of Hepatobiliary Surgery and Liver Transplantation, Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Matías A Avila
- Hepatology Laboratory, Solid Tumors Program, CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), CIMA, University of Navarra, Instituto de Salud Carlos III, Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain
| | - Manuel Rodríguez-Perálvarez
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
- Department of Hepatology and Liver Transplantation, Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Raúl M Luque
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Juan L López-Cánovas
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Manuel D Gahete
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain.
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain.
| |
Collapse
|
19
|
Long H, Zhou J, Zhou C, Xie S, Wang J, Tan M, Xu J. Proteomic Characterization of Liver Cancer Cells Treated with Clinical Targeted Drugs for Hepatocellular Carcinoma. Biomedicines 2025; 13:152. [PMID: 39857736 PMCID: PMC11760458 DOI: 10.3390/biomedicines13010152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/30/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Hepatocellular carcinoma (HCC) remains a significant global health concern, primarily due to the limited efficacy of targeted therapies, which are often compromised by drug resistance and adverse side effects. Methods: In this study, we utilized a Tandem Mass Tag (TMT)-based quantitative proteomic approach to analyze global protein expression and serine/threonine/tyrosine (S/T/Y) phosphorylation modifications in HepG2 cells following treatment with three clinically relevant hepatocellular carcinoma-targeted agents: apatinib, regorafenib, and lenvatinib. Results: Utilizing KEGG pathway enrichment analysis, biological process enrichment analysis, and protein interaction network analysis, we elucidated the common and specific metabolic pathways, biological processes, and protein interaction regulatory networks influenced by three liver cancer therapeutics. The study additionally proposed potential combinational treatment strategies, highlighting a possible synergistic interaction between HCC-targeted drugs and the DNA methyltransferase inhibitor. Furthermore, through the integration of clinical phosphorylation site data, we identified several phosphorylation sites that exhibited higher abundance in tumor tissues compared to adjacent non-tumor tissues. These sites were associated with poor prognosis and elevated functional scores. Conclusions: In summary, this study conducted an in-depth analysis of the molecular alterations in proteins and phosphorylation modifications induced by clinical HCC-targeted drugs, predicting drug combination strategies and therapeutic targets.
Collapse
Affiliation(s)
- Hezhou Long
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (H.L.); (J.Z.)
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China;
| | - Jiafu Zhou
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (H.L.); (J.Z.)
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China;
| | - Changxia Zhou
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; (C.Z.); (S.X.)
| | - Shuyu Xie
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; (C.Z.); (S.X.)
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingling Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China;
| | - Minjia Tan
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (H.L.); (J.Z.)
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China;
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; (C.Z.); (S.X.)
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Junyu Xu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (H.L.); (J.Z.)
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China;
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; (C.Z.); (S.X.)
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
20
|
Chen A, Yin K, Liu Y, Hu L, Cui Q, Wan X, Yang W. WEE Family Kinase Inhibitors Combined with Sorafenib Can Selectively Inhibit HCC Cell Proliferation. Curr Cancer Drug Targets 2025; 25:370-385. [PMID: 38860904 DOI: 10.2174/0115680096298370240520093003] [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: 01/08/2024] [Revised: 04/10/2024] [Accepted: 04/24/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Sorafenib is currently the first choice for the treatment of patients with advanced hepatocellular carcinoma, but its therapeutic effect is still limited. OBJECTIVES This study aims to examine whether WEE family kinase inhibitors can enhance the anticancer effect of sorafenib. METHODS We analyzed the expression levels of PKMYT1 kinase and WEE1 kinase in HCC, studied the inhibitory effect of PKMYT1 kinase inhibitor RP-6306, WEE1 kinase inhibitor adavosertib combined with sorafenib on the proliferation of HCC cells, and detected the effect of drug combination on CDK1 phosphorylation. RESULTS We found that PKMYT1 and WEE1 were upregulated in HCC and were detrimental to patient survival. Cell experiments showed that both RP-6306 and adavosertib (1-100 μM) inhibited the proliferation of HCC cell lines in a dose-dependent manner alone, and the combination of the two drugs had a synergistic effect. In HCC cell lines, sorafenib combined with RP-6306 or adavosertib showed a synergistic antiproliferation effect and less toxicity to normal cells. Sorafenib combined with RP-6306 and adavosertib further inhibited the proliferation of HCC cells and caused complete dephosphorylation of CDK1. CONCLUSION Taken together, our findings provide experimental evidence for the future use of sorafenib in combination with RP-6306 or adavosertib for the treatment of HCC.
Collapse
Affiliation(s)
- Anling Chen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, 230031, China
| | - Ke Yin
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Yu Liu
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Lei Hu
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Qianwen Cui
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, 230031, China
| | - Xiaofeng Wan
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Wulin Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| |
Collapse
|
21
|
Wang L, Zhao Z, Shu K, Ma M. MPCD Index for Hepatocellular Carcinoma Patients Based on Mitochondrial Function and Cell Death Patterns. Int J Mol Sci 2024; 26:118. [PMID: 39795978 PMCID: PMC11719604 DOI: 10.3390/ijms26010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/24/2024] [Accepted: 12/24/2024] [Indexed: 01/30/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous cancer with a poor prognosis. During the development of cancer cells, mitochondria influence various cell death patterns by regulating metabolic pathways such as oxidative phosphorylation. However, the relationship between mitochondrial function and cell death patterns in HCC remains unclear. In this study, we used a comprehensive machine learning framework to construct a mitochondrial functional activity-associated programmed cell death index (MPCDI) based on scRNA-seq and RNA-seq data from TCGA, GEO, and ICGC datasets. The index signature was used to classify HCC patients, and studied the multi-omics features, immune microenvironment, and drug sensitivity of the subtypes. Finally, we constructed the MPCDI signature consisting of four genes (S100A9, FYN, LGALS3, and HMOX1), which was one of the independent risk factors for the prognosis of HCC patients. The HCC patients were divided into high- and low-MPCDI groups, and the immune status was different between the two groups. Patients with a high MPCDI had higher TIDE scores and poorer responses to immunotherapy, suggesting that high-MPCDI patients might not be suitable for immunotherapy. By analyzing the drug sensitivity data of CTRP, GDSC, and PRISM databases, it was found that staurosporine has potential therapeutic significance for patients with a high MPCDI. In summary, based on the characteristics of mitochondria function and PCD patterns, we used single-cell and transcriptome data to identify four genes and construct the MPCDI signature, which provided new perspectives and directions for the clinical diagnosis and personalized treatment of HCC patients.
Collapse
Affiliation(s)
- Longxing Wang
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, China; (L.W.); (Z.Z.); (K.S.)
| | - Zhiming Zhao
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, China; (L.W.); (Z.Z.); (K.S.)
| | - Kunxian Shu
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, China; (L.W.); (Z.Z.); (K.S.)
| | - Mingyue Ma
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, China; (L.W.); (Z.Z.); (K.S.)
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
22
|
Das D, Wang X, Chiu YC, Bouamar H, Sharkey FE, Lopera JE, Lai Z, Weintraub ST, Han X, Zou Y, Chen HIH, Zeballos Torrez CR, Gu X, Cserhati M, Michalek JE, Halff GA, Chen Y, Zheng S, Cigarroa FG, Sun LZ. Integrative multi-omics characterization of hepatocellular carcinoma in Hispanic patients. J Natl Cancer Inst 2024; 116:1961-1978. [PMID: 39189979 PMCID: PMC11630563 DOI: 10.1093/jnci/djae207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/23/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND The incidence and mortality rates of hepatocellular carcinoma among Hispanic individuals in the United States are much higher than in non-Hispanic White people. We conducted multi-omics analyses to elucidate molecular alterations in hepatocellular carcinoma among Hispanic patients. METHODS Paired tumor and adjacent nontumor samples were collected from 31 Hispanic hepatocellular carcinomas in South Texas for genomic, transcriptomic, proteomic, and metabolomic profiling. Serum lipids were profiled in 40 Hispanic and non-Hispanic patients with or without clinically diagnosed hepatocellular carcinoma. RESULTS Exome sequencing revealed high mutation frequencies of AXIN2 and CTNNB1 in South Texas Hispanic hepatocellular carcinoma patients, suggesting a predominant activation of the Wnt/β-catenin pathway. TERT promoter mutations were also statistically significantly more frequent in the Hispanic cohort (Fisher exact test, P < .05). Cell cycles and liver function were positively and negatively enriched, respectively, with gene set enrichment analysis. Gene sets representing specific liver metabolic pathways were associated with dysregulation of corresponding metabolites. Negative enrichment of liver adipogenesis and lipid metabolism corroborated with a significant reduction in most lipids in serum samples of hepatocellular carcinoma patients (paired t test, P < .0001). Two hepatocellular carcinoma subtypes from our Hispanic cohort were identified and validated with the Cancer Genome Atlas liver cancer cohort. Patients with better overall survival showed higher activity of immune and angiogenesis signatures and lower activity of liver function-related gene signatures. They also had higher levels of immune checkpoint and immune exhaustion markers. CONCLUSIONS Our study revealed specific molecular features of Hispanic hepatocellular carcinoma and potential biomarkers for therapeutic management. It provides a unique resource for studying Hispanic hepatocellular carcinoma.
Collapse
Affiliation(s)
- Debodipta Das
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xiaojing Wang
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yu-Chiao Chiu
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hakim Bouamar
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Francis E Sharkey
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jorge E Lopera
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Zhao Lai
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Susan T Weintraub
- Department of Biochemistry & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yi Zou
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hung-I H Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Carla R Zeballos Torrez
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xiang Gu
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Matyas Cserhati
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Joel E Michalek
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Glenn A Halff
- Transplant Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Siyuan Zheng
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Francisco G Cigarroa
- Transplant Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Lu-Zhe Sun
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
23
|
Alvarado-Tapias E, Maya-Miles D, Albillos A, Aller R, Ampuero J, Andrade RJ, Arechederra M, Aspichueta P, Banales JM, Blas-García A, Caparros E, Cardoso Delgado T, Carrillo-Vico A, Claria J, Cubero FJ, Díaz-Ruiz A, Fernández-Barrena MG, Fernández-Iglesias A, Fernández-Veledo S, Francés R, Gallego-Durán R, Gracia-Sancho J, Irimia M, Lens S, Martínez-Chantar ML, Mínguez B, Muñoz-Hernández R, Nogueiras R, Ramos-Molina B, Riveiro-Barciela M, Rodríguez-Perálvarez ML, Romero-Gómez M, Sabio G, Sancho-Bru P, Ventura-Cots M, Vidal S, Gahete MD. Proceedings of the 5th Meeting of Translational Hepatology, organized by the Spanish Association for the Study of the Liver (AEEH). GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:502207. [PMID: 38723772 DOI: 10.1016/j.gastrohep.2024.502207] [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: 04/15/2024] [Accepted: 05/02/2024] [Indexed: 11/30/2024]
Abstract
This is the summary report of the 5th Translational Hepatology Meeting, endorsed by the Spanish Association for the Study of the Liver (AEEH) and held in Seville, Spain, in October 2023. The meeting aimed to provide an update on the latest advances in the field of basic and translational hepatology, covering different molecular, cellular, and pathophysiological aspects of the most relevant clinical challenges in liver pathologies. This includes the identification of novel biomarkers and diagnostic tools, the understanding of the relevance of immune response and inflammation in liver diseases, the characterization of current medical approaches to reverse liver diseases, the incorporation of novel molecular insights through omics techniques, or the characterization of the impact of toxic and metabolic insults, as well as other organ crosstalk, in liver pathophysiology.
Collapse
Affiliation(s)
- Edilmar Alvarado-Tapias
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Gastroenterology, Hospital Santa Creu I Sant Pau, Institut de Recerca Sant Pau, Universidad Autónoma de Barcelona, Barcelona, Spain.
| | - Douglas Maya-Miles
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain.
| | - Agustin Albillos
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Servicio de Gastroenterología y Hepatología, Hospital Universitario Ramón y Cajal/Universidad de Alcalá/Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Rocio Aller
- BioCritic, Group for Biomedical Research in Critical Care Medicine, Spain; Department of Medicine, Dermatology and Toxicology, Universidad de Valladolid, Spain; Gastroenterology Unit, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Javier Ampuero
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain
| | - Raul J Andrade
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Maria Arechederra
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain; Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain
| | - Patricia Aspichueta
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain; Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Jesus M Banales
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute - Donostia University Hospital - University of the Basque Country (UPV/EHU), Ikerbasque, Donostia-San Sebastian, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Ana Blas-García
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Departamento de Fisiología, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Av. de Catalunya, 21, 46020 Valencia, Spain
| | - Esther Caparros
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Grupo de Inmunobiología Hepática e Intestinal, Departamento Medicina Clínica, Universidad Miguel Hernández, San Juan, Spain; Instituto de Investigación Sanitaria ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
| | - Teresa Cardoso Delgado
- Biobizkaia Health Research Institute, Barakaldo, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Antonio Carrillo-Vico
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Joan Claria
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Biochemistry and Molecular Genetics Service, Hospital Clínic, IDIBAPS, Barcelona, Spain; University of Barcelona, Spain
| | - Francisco Javier Cubero
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), Madrid, Spain
| | - Alberto Díaz-Ruiz
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Maite G Fernández-Barrena
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain; Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Spain
| | - Anabel Fernández-Iglesias
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Vascular Biology Research Group, IDIBAPS, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Ruben Francés
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Grupo de Inmunobiología Hepática e Intestinal, Departamento Medicina Clínica, Universidad Miguel Hernández, San Juan, Spain; Instituto de Investigación Sanitaria ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
| | - Rocío Gallego-Durán
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain
| | - Jordi Gracia-Sancho
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Vascular Biology Research Group, IDIBAPS, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Manuel Irimia
- Universitat Pompeu Fabra (UPF), Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, ICREA, Barcelona, Spain
| | - Sabela Lens
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Liver Unit, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - María Luz Martínez-Chantar
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
| | - Beatriz Mínguez
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Rocío Muñoz-Hernández
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain; Departamento de fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Rubén Nogueiras
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain; Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain
| | - Bruno Ramos-Molina
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Mar Riveiro-Barciela
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Manuel L Rodríguez-Perálvarez
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Hepatology and Liver Transplantation, Reina Sofia University Hospital, Cordoba, Spain; Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Cordoba, Spain
| | - Manuel Romero-Gómez
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Stress Kinases in Diabetes, Cancer and Biochemistry, Spain; Centro Nacional de Investigaciones Oncologicas (CNIO), Organ Crosstalk in Metabolic Diseases, Madrid, Spain
| | - Pau Sancho-Bru
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Meritxell Ventura-Cots
- Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; Center for Liver Diseases, Pittsburgh Liver Research Center, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Silvia Vidal
- Group of Inflammatory Diseases, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Manuel D Gahete
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Spain; Molecular Hepatology Group, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Spain; Reina Sofia University Hospital, Cordoba, Spain.
| |
Collapse
|
24
|
Kaldjob-Heinrich L, Nuciforo S, Lemke S, Stahl A, Czemmel S, Babaei S, Blukacz L, Meier MA, Zhang Y, Schürch CM, Gonzalez-Menendez I, Woelffing P, Malek NP, Scheble V, Nahnsen S, Claassen M, Templin M, Bösmüller H, Heim MH, Dauch D, Bitzer M. Adenosine Receptor 3 in Liver Cancer: Expression Variability, Epigenetic Modulation, and Enhanced Histone Deacetylase Inhibitor Effects. GASTRO HEP ADVANCES 2024; 4:100590. [PMID: 39911497 PMCID: PMC11795062 DOI: 10.1016/j.gastha.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/13/2024] [Indexed: 02/07/2025]
Abstract
Background and Aims Primary liver cancer, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), has low response rates to existing treatments, highlighting the urgent need for novel treatment options. Adenosine A3 receptor (ADORA3) signaling has emerged as a potential target. Namodenoson, an ADORA3 agonist, has shown promise in early clinical trials for HCC. However, further data are required to clarify ADORA3 expression patterns in liver cancer, mechanisms of action, and the potential for combination therapies to inform patient selection for future clinical trials. Methods Patient-derived tissue microarrays and RNA-sequencing were employed to investigate ADORA3 expression. Cellular responses to ADORA3 stimulation and combination treatments were studied in HCC and CCA cell lines and patient-derived organoids (PDOs). Genome-wide RNA-Seq analysis, mRNA analysis, and DigiWest protein profiling were performed. Results Tissue microarray analysis revealed higher ADORA3 expression in nonmalignant samples and a subset of tumors with weak or absent ADORA3 expression. This was supported by RNA sequencing data from The Cancer Genome Atlas and needle biopsy samples. Cell lines and PDOs exhibited antiproliferative effects with the ADORA3 agonist Namodenoson, confirmed by receptor dependency tests with specific antagonists and siRNA experiments. Genome-wide RNA-Seq analysis suggested chromatin remodeling events after ADORA3 stimulation. mRNA expression and DigiWest profiling identified downregulation of histone deacetylases and histone H3 modifications. Combination treatments with different ADORA3 agonists and histone deacetylase inhibitors significantly enhanced antiproliferative effects in almost all selected combinations, supported by investigations in PDOs. Conclusion ADORA3 expression varies considerably in HCC or CCA, ranging from high to absent receptor detection. This observation might help to identify patients for clinical studies. Additionally, Namodenoson's epigenetic modulating activity suggests epigenetic drugs as promising candidates for combination treatment.
Collapse
Affiliation(s)
| | - Sandro Nuciforo
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
- Clinic of Gastroenterology and Hepatology, Clarunis University Center for Gastrointestinal and Liver Diseases Basel, Basel, Switzerland
| | - Steffen Lemke
- Quantitative Biology Center (QBiC), Eberhard-Karls University, Tuebingen, Germany
- M3-Research Center for Malignome, Metabolome and Microbiome, Eberhard-Karls University, Tuebingen, Germany
| | - Aaron Stahl
- NMI, Natural and Medical Sciences Institute at the University of Tuebingen, Reutlingen, Germany
| | - Stefan Czemmel
- Quantitative Biology Center (QBiC), Eberhard-Karls University, Tuebingen, Germany
- M3-Research Center for Malignome, Metabolome and Microbiome, Eberhard-Karls University, Tuebingen, Germany
| | - Sepideh Babaei
- Department Internal Medicine I, Eberhard-Karls University, Tuebingen, Germany
- M3-Research Center for Malignome, Metabolome and Microbiome, Eberhard-Karls University, Tuebingen, Germany
| | - Lauriane Blukacz
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Marie-Anne Meier
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
- Clinic of Gastroenterology and Hepatology, Clarunis University Center for Gastrointestinal and Liver Diseases Basel, Basel, Switzerland
| | - Yizheng Zhang
- Department of Pathology and Neuropathology, Eberhard Karls University, Tübingen, Germany
| | - Christian M. Schürch
- Department of Pathology and Neuropathology, Eberhard Karls University, Tübingen, Germany
- iFIT Cluster of Excellence EXC 2180 ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard-Karls University, Tuebingen, Germany
| | - Irene Gonzalez-Menendez
- Department of Pathology and Neuropathology, Eberhard Karls University, Tübingen, Germany
- iFIT Cluster of Excellence EXC 2180 ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard-Karls University, Tuebingen, Germany
| | - Pascal Woelffing
- iFIT Cluster of Excellence EXC 2180 ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard-Karls University, Tuebingen, Germany
- Department of Medical Oncology and Pneumology, Eberhard-Karls University, Tuebingen, Germany
| | - Nisar P. Malek
- Department Internal Medicine I, Eberhard-Karls University, Tuebingen, Germany
- M3-Research Center for Malignome, Metabolome and Microbiome, Eberhard-Karls University, Tuebingen, Germany
- iFIT Cluster of Excellence EXC 2180 ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard-Karls University, Tuebingen, Germany
- Center for Personalized Medicine, Eberhard-Karls University, Tuebingen, Germany
| | - Veit Scheble
- Department Internal Medicine I, Eberhard-Karls University, Tuebingen, Germany
| | - Sven Nahnsen
- Quantitative Biology Center (QBiC), Eberhard-Karls University, Tuebingen, Germany
- M3-Research Center for Malignome, Metabolome and Microbiome, Eberhard-Karls University, Tuebingen, Germany
| | - Manfred Claassen
- Department Internal Medicine I, Eberhard-Karls University, Tuebingen, Germany
- M3-Research Center for Malignome, Metabolome and Microbiome, Eberhard-Karls University, Tuebingen, Germany
- Department of Computer Science, University of Tübingen, Tübingen, Germany
- Machine Learning in Science, Excellence Cluster Machine Learning, University of Tübingen, Tübingen, Germany
| | - Markus Templin
- NMI, Natural and Medical Sciences Institute at the University of Tuebingen, Reutlingen, Germany
| | - Hans Bösmüller
- Department of Pathology and Neuropathology, Eberhard Karls University, Tübingen, Germany
| | - Markus H. Heim
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
- Clinic of Gastroenterology and Hepatology, Clarunis University Center for Gastrointestinal and Liver Diseases Basel, Basel, Switzerland
| | - Daniel Dauch
- iFIT Cluster of Excellence EXC 2180 ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard-Karls University, Tuebingen, Germany
- Department of Medical Oncology and Pneumology, Eberhard-Karls University, Tuebingen, Germany
| | - Michael Bitzer
- Department Internal Medicine I, Eberhard-Karls University, Tuebingen, Germany
- M3-Research Center for Malignome, Metabolome and Microbiome, Eberhard-Karls University, Tuebingen, Germany
- iFIT Cluster of Excellence EXC 2180 ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard-Karls University, Tuebingen, Germany
- Center for Personalized Medicine, Eberhard-Karls University, Tuebingen, Germany
| |
Collapse
|
25
|
Su RY, Xu CH, Guo HJ, Meng LJ, Zhuo JY, Xu N, Li HG, He CY, Zhang XY, Lian ZX, Wang S, Cao C, Zhou R, Lu D, Zheng SS, Wei XY, Xu X. Oncogenic cholesterol rewires lipid metabolism in hepatocellular carcinoma via the CSNK2A1-IGF2R Ser2484 axis. J Adv Res 2024:S2090-1232(24)00540-X. [PMID: 39547439 DOI: 10.1016/j.jare.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 10/02/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024] Open
Abstract
INTRODUCTION Alcohol consumption and hepatitis B virus (HBV) infection are common risk factors for hepatocellular carcinoma (HCC). However, few studies have focused on elucidating the mechanisms of HCC with combined alcohol and HBV etiology. OBJECTIVES We aimed to investigate the molecular features of alcohol and HBV on HCC and to seek out potential therapeutic strategies. METHODS Two independent cohorts of HCC patients (n = 539 and n = 140) were included to investigate HCC with synergetic alcohol and HBV (AB-HCC) background. Patient-derived cell lines, organoids, and xenografts were used to validate the metabolic fragile. High-throughput drug screening (1181 FDA-approved anticancer drugs) was leveraged to explore the potential therapeutic agents. RESULTS Here, we delineated AB-HCC as a distinctive metabolic subtype, hallmarked by oncogenic cholesterol, through the integration of clinical cohorts, proteomics, phosphoproteomics, and spatial transcriptome. Mechanistically, our findings revealed that cholesterol directly binds to CSNK2A1 (Casein Kinase 2 Alpha 1), augmenting its kinase activity and leading to phosphorylation of IGF2R (Insulin-Like Growth Factor 2 Receptor) at Ser2484. This cascade rewires lipid-driven mitochondrial oxidative phosphorylation, spawns reactive oxygen species measured by malondialdehyde assay, and perpetuates a positive feedback loop for cholesterol biosynthesis, ultimately culminating in tumorigenesis. Initial transcriptional activation of CSNK2A1 is driven by upregulation of RAD21 in AB-HCC. Our cholesterol profiling exposes AB-HCC's compensatory mechanism of AB-HCC, which capitalizes on both uptake and biosynthesis of cholesterol to confer survival edge. Moreover, high-throughput drug screening coupled with in vivo validation has uncovered the susceptibilities of AB-HCC, which can be effectively addressed by a combination of dietary cholesterol restriction and oral administration of Fostamatinib. The CSNK2A1-mediated cholesterol biosynthesis pathway has been implicated in various cancers characterized by cholesterol metabolism. CONCLUSION These findings not only pinpoint the oncogenic metabolite cholesterol as a hidden culprit in AB-HCC subtype, but also enlighten a novel combination strategy to rejuvenate tumor metabolism.
Collapse
Affiliation(s)
- Ren-Yi Su
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Chen-Hao Xu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Hai-Jun Guo
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China; Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Hangzhou 310006, China
| | - Li-Jun Meng
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou 310006, China
| | - Jian-Yong Zhuo
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Hangzhou 310006, China; Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou 310006, China
| | - Nan Xu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Hui-Gang Li
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Chi-Yu He
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xuan-Yu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
| | - Zheng-Xin Lian
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Hangzhou 310006, China; Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou 310006, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou 310006, China
| | - Chenhao Cao
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, and College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| | - Di Lu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310059, China.
| | - Shu-Sen Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China; Department of Hepatobiliary and Pancreatic Surgery, Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310022, China.
| | - Xu-Yong Wei
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Hangzhou 310006, China; Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou 310006, China.
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310059, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China.
| |
Collapse
|
26
|
Garcia CJC, Grisetti L, Tiribelli C, Pascut D. The ncRNA-AURKA Interaction in Hepatocellular Carcinoma: Insights into Oncogenic Pathways, Therapeutic Opportunities, and Future Challenges. Life (Basel) 2024; 14:1430. [PMID: 39598228 PMCID: PMC11595987 DOI: 10.3390/life14111430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/15/2024] [Accepted: 11/03/2024] [Indexed: 11/29/2024] Open
Abstract
Hepatocellular carcinoma (HCC) represents a major public health concern and ranks among the leading cancer-related mortalities globally. Due to the frequent late-stage diagnosis of HCC, therapeutic options remain limited. Emerging evidence highlights the critical role of non-coding RNAs (ncRNAs) in the regulation of Aurora kinase A (AURKA), one of the key hub genes involved in several key cancer pathways. Indeed, the dysregulated interaction between ncRNAs and AURKA contributes to tumor development, progression, and therapeutic resistance. This review delves into the interplay between ncRNAs and AURKA and their role in hepatocarcinogenesis. Recent findings underscore the involvement of the ncRNAs and AURKA axis in tumor development and progression. Furthermore, this review also discusses the clinical significance of targeting ncRNA-AURKA axes, offering new perspectives that could lead to innovative therapeutic strategies aimed at improving outcomes for HCC patients.
Collapse
Affiliation(s)
- Clarissa Joy C. Garcia
- Liver Cancer Unit, Fondazione Italiana Fegato—ONLUS, 34149 Trieste, Italy
- Department of Life Sciences, Università degli Studi di Trieste, 34127 Trieste, Italy
| | - Luca Grisetti
- National Institute of Gastroenterology—IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy
| | - Claudio Tiribelli
- Liver Cancer Unit, Fondazione Italiana Fegato—ONLUS, 34149 Trieste, Italy
| | - Devis Pascut
- Liver Cancer Unit, Fondazione Italiana Fegato—ONLUS, 34149 Trieste, Italy
| |
Collapse
|
27
|
Creighton CJ. Clinical proteomics towards multiomics in cancer. MASS SPECTROMETRY REVIEWS 2024; 43:1255-1269. [PMID: 36495097 DOI: 10.1002/mas.21827] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Recent technological advancements in mass spectrometry (MS)-based proteomics technologies have accelerated its application to study greater and greater numbers of human tumor specimens. Over the last several years, the Clinical Proteomic Tumor Analysis Consortium, the International Cancer Proteogenome Consortium, and others have generated MS-based proteomic profiling data combined with corresponding multiomics data on thousands of human tumors to date. Proteomic data sets in the public domain can be re-examined by other researchers with different questions in mind from what the original studies explored. In this review, we examine the increasing role of proteomics in studying cancer, along with the potential for previous studies and their associated data sets to contribute to improving the diagnosis and treatment of cancer in the clinical setting. We also explore publicly available proteomics and multi-omics data from cancer cell line models to show how such data may aid in identifying therapeutic strategies for cancer subsets.
Collapse
Affiliation(s)
- Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
28
|
Jiang H, Cannella R, Wu Z, Beaufrère A, Dioguardi Burgio M, Sartoris R, Wang Y, Qin Y, Chen J, Chen Y, Chen W, Shi Y, Song B, Ronot M. Prognostic Implications of MRI-assessed Intratumoral Fat in Hepatocellular Carcinoma: An Asian and European Cohort Study. Radiology 2024; 313:e233471. [PMID: 39499179 DOI: 10.1148/radiol.233471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Background The clinicopathologic-radiologic and prognostic characteristics of intratumoral fat in hepatocellular carcinoma (HCC) are critical for personalized treatment but remain understudied. Purpose To investigate the clinicopathologic-radiologic associations and prognostic implications of MRI-assessed intratumoral fat in HCCs. Materials and Methods This retrospective cohort study included consecutive adult patients who underwent resection for solitary HCCs and preoperative contrast-enhanced MRI from two tertiary-care hospitals in East Asia (March 2011 to December 2021) and Western Europe (September 2012 to December 2019). MRI scans were independently evaluated by three radiologists at each hospital. Based on Liver Imaging Reporting and Data System (LI-RADS) version 2018, intratumoral fat was defined as "fat in mass more than adjacent liver," and the homogeneous subtype was defined as intratumoral fat "in absence of mosaic and nodule-in-nodule architecture." Recurrence-free survival (RFS) and overall survival (OS) were estimated using the Kaplan-Meier method and compared using the log-rank test. Cox regression analyses were conducted to identify factors associated with RFS and OS. Results A total of 933 patients were included in the Asian (n = 736; median age, 53 years [IQR, 45-62 years]; 626 male) and European (n = 207; median age, 64 years [IQR, 55-70 years]; 161 male) cohorts. MRI-assessed intratumoral fat was detected in 30% (215 of 726) and 31% (64 of 207) of patients in the Asian and European cohorts, respectively (P = .72). In both cohorts, the steatohepatitic subtype, nonperipheral washout, enhancing capsule, and mosaic architecture were more frequent in tumors with intratumoral fat (P value range, <.001 to .04). Intratumoral fat in general was not associated with RFS or OS in either cohort (P value range, .48-.97). However, in the Asian cohort, homogeneous intratumoral fat was associated with longer RFS (hazard ratio [HR], 0.60; P = .009) and OS (HR, 0.33; P = .008) in multivariable Cox regression analyses. Conclusion MRI-assessed intratumoral fat was more frequent in steatohepatitic HCCs and associated with nonperipheral washout, enhancing capsule, and mosaic architecture. Although intratumoral fat was generally nonprognostic, homogeneous intratumoral fat was associated with longer RFS and OS in the Asian cohort. Published under a CC BY 4.0 license. Supplemental material is available for this article. See also the editorial by Harmath in this issue.
Collapse
Affiliation(s)
- Hanyu Jiang
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Roberto Cannella
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Zhenru Wu
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Aurélie Beaufrère
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Marco Dioguardi Burgio
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Riccardo Sartoris
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Yanshu Wang
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Yun Qin
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Jie Chen
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Yidi Chen
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Weixia Chen
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Yujun Shi
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Bin Song
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| | - Maxime Ronot
- From the Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China (H.J., Y.W., Y.Q., J.C., Y.C., W.C., B.S.); Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy 92110, France (R.C., M.D.B., R.S., M.R.); Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy (R.C.); Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China (Z.W., Y.S.); Université Paris CIté, CRI, INSERM UMR 1149, Paris & Department of Pathology, FHU MOSAIC, AP-HP.Nord, Beaujon Hospital, Clichy, France (A.B.); and Department of Radiology, Sanya People's Hospital, Sanya, China (B.S.)
| |
Collapse
|
29
|
Albadawi H, Zhang Z, Keum H, Cevik E, Nagalo BM, Gunduz S, Kita H, Oklu R. Percutaneous Delivery of Oncogel for Targeted Liver Tumor Ablation and Controlled Release of Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406080. [PMID: 39148179 PMCID: PMC11543513 DOI: 10.1002/adma.202406080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/30/2024] [Indexed: 08/17/2024]
Abstract
Advanced-stage liver cancers are associated with poor prognosis and have limited treatment options, often leading the patient to hospice care. Percutaneous intratumoral injection of anticancer agents has emerged as a potential alternative to systemic therapy to overcome tumor barriers, increase bioavailability, potentiate immunotherapy, and avoid systemic toxicity, which advanced-stage cancer patients cannot tolerate. Here, an injectable OncoGel (OG) comprising of a nanocomposite hydrogel loaded with an ionic liquid (IL) is developed for achieving a predictable and uniform tumor ablation and long-term slow release of anticancer agents into the ablation zone. Rigorous mechanical, physiochemical, drug release, cytotoxicity experiments, and ex vivo human tissue testing identify an injectable version of the OG with bactericidal properties against highly resistant bacteria. Intratumoral injection of OG loaded with Nivolumab (Nivo) and doxorubicin (Dox) into highly malignant tumor models in mice, rats, and rabbits demonstrates enhanced survival and tumor regression associated with robust tissue ablation and drug distribution throughout the tumor. Mass cytometry and proteomic studies in a mouse model of colorectal cancer that often metastasizes to the liver indicate an enhanced anticancer immune response following the intratumoral injection of OG. OG may augment immunotherapy and potentially improve outcomes in liver cancer patients.
Collapse
Affiliation(s)
- Hassan Albadawi
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Zefu Zhang
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Hyeongseop Keum
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Enes Cevik
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Bolni M Nagalo
- University of Arkansas for Medical Sciences, College of Medicine, Department of Pathology, 301 West Markham Street, Little Rock, AR, 72205, USA
| | - Seyda Gunduz
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
- Department of Medical Oncology, Istinye University; Bahcesehir Liv Hospital, Istanbul 34517, Turkey
| | - Hirohito Kita
- Department of Immunology, Division of Allergy, Asthma, and Clinical Immunology and the Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona 85259, USA
| | - Rahmi Oklu
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| |
Collapse
|
30
|
Fan M, Hu J, Xu X, Chen J, Zhang W, Zheng X, Pan J, Xu W, Feng S. Mass spectrometry-based multi-omics analysis reveals distinct molecular features in early and advanced stages of hepatocellular carcinoma. Heliyon 2024; 10:e38182. [PMID: 39381095 PMCID: PMC11456867 DOI: 10.1016/j.heliyon.2024.e38182] [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: 02/04/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
Hepatocellular Carcinoma (HCC) is a serious primary solid tumor that is prevalent worldwide. Due to its high mortality rate, it is crucial to explore both early diagnosis and advanced treatment for HCC. In recent years, multi-omics approaches have emerged as promising tools to identify biomarkers and investigate molecular mechanisms of biological processes and diseases. In this study, we performed proteomics, phosphoproteomics, metabolomics, and lipidomics to reveal the molecular features of early- and advanced-stage HCC. The data obtained from these omics were analyzed separately and then integrated to provide a comprehensive understanding of the disease. The multi-omics results unveiled intricate biological pathways and interaction networks underlying the initiation and progression of HCC. Moreover, we proposed specific potential biomarker panels for both early- and advanced-stage HCC by overlapping our data with CPTAC database for HCC diagnosis, and deduced novel insights and mechanisms related to HCC origination and development, such as glucose depletion during tumor progression, ROCK1 deactivation and GSK3A activation.
Collapse
Affiliation(s)
- Mingzhu Fan
- Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou, 310024, Zhejiang, China
- Mass Spectrometry & Metabolomics Core Facility, The Biomedical Research Core Facility, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Jin Hu
- Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou, 310024, Zhejiang, China
- Mass Spectrometry & Metabolomics Core Facility, The Biomedical Research Core Facility, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Xiaoyan Xu
- Mass Spectrometry & Metabolomics Core Facility, The Biomedical Research Core Facility, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Jia Chen
- Mass Spectrometry & Metabolomics Core Facility, The Biomedical Research Core Facility, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Wenwen Zhang
- Mass Spectrometry & Metabolomics Core Facility, The Biomedical Research Core Facility, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Xiaoping Zheng
- Pathology Department, Shulan (Hangzhou) Hospital, Hangzhou, 311112, Zhejiang, China
| | - Jinheng Pan
- Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Wei Xu
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Hangzhou Tongchuang Medical Laboratory, Shulan Health Group, Hangzhou, 310015, Zhejiang, China
| | - Shan Feng
- Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou, 310024, Zhejiang, China
- Mass Spectrometry & Metabolomics Core Facility, The Biomedical Research Core Facility, Westlake University, Hangzhou, 310024, Zhejiang, China
| |
Collapse
|
31
|
Huang Y, Peng M, Yu W, Li H. Activation of Wnt/β-catenin signaling promotes immune evasion via the β-catenin/IKZF1/CCL5 axis in hepatocellular carcinoma. Int Immunopharmacol 2024; 138:112534. [PMID: 38941667 DOI: 10.1016/j.intimp.2024.112534] [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: 05/08/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Immune checkpoint therapy (ICT) has been shown to produce durable responses in various cancer patients. However, its efficacy is notably limited in hepatocellular carcinoma (HCC), with only a small percentage of patients responding positively to treatment. The mechanism underlying resistance to ICT in HCC remains poorly understood. Here, we showed that combination treatment of ICG-001, an inhibitor of the Wnt/β-catenin signaling pathway, with anti-PD-1 antibody effectively suppresses tumor growth and promotes the infiltration of immune cells such as DCs and CD8+ T cells in the tumor microenvironment (TME). By inhibiting the activity of β-catenin and blocking its binding to the transcription factor IKAROS family zinc finger 1 (IKZF1), ICG-001 upregulated the expression of CCL5. Moreover, IKZF1 regulated the activity of the CCL5 promoter and its endogenous expression. Through inhibition of the WNT/β-catenin signaling pathway, upregulation of the expression of CCL5 was achieved, which subsequently recruited more DCs into the TME via C-C motif chemokine receptor 5 (CCR5). This, in turn, resulted in an increase in the infiltration of CD8+ T cells in the TME, thereby enhancing the antitumor immune response. Analysis of a tissue microarray derived from HCC patient samples revealed a positive correlation between survival rate and prognosis and the expression levels of CCL5/CD8. In conclusion, our findings suggest that combined application of ICG-001 and anti-PD-1 antibody exhibits significantly enhanced antitumor efficacy. Hence, combining a WNT/β-catenin signaling pathway inhibitor with anti-PD-1 therapy may be a promising treatment strategy for patients with HCC.
Collapse
Affiliation(s)
- Yamei Huang
- Department of Pathology and Pathophysiology, Medical School of Southeast University, China
| | - Min Peng
- Department of Pathogenic Biology and Immunology, Medical School of Southeast University, China
| | - Weiping Yu
- Department of Pathology and Pathophysiology, Medical School of Southeast University, China.
| | - Hui Li
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, China.
| |
Collapse
|
32
|
Xie P, Yin Q, Wang S, Song D. Prognostic Protein Biomarker Screening for Thyroid Carcinoma Based on Cancer Proteomics Profiles. Biomedicines 2024; 12:2066. [PMID: 39335579 PMCID: PMC11428938 DOI: 10.3390/biomedicines12092066] [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: 07/25/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Thyroid carcinoma (THCA) ranks among the most prevalent cancers globally. Integrating advanced genomic and proteomic analyses to construct a protein-based prognostic model promises to identify effective biomarkers and explore new therapeutic avenues. In this study, proteomic data from The Cancer Proteomics Atlas (TCPA) and clinical data from The Cancer Genome Atlas (TCGA) were utilized. Using Kaplan-Meier, Cox regression, and LASSO penalized Cox analyses, we developed a prognostic risk model comprising 13 proteins (S100A4, PAI1, IGFBP2, RICTOR, B7-H3, COLLAGENVI, PAR, SNAIL, FAK, Connexin-43, Rheb, EVI1, and P90RSK_pT359S363). The protein prognostic model was validated as an independent predictor of survival time in THCA patients, based on risk curves, survival analysis, receiver operating characteristic curves and independent prognostic analysis. Additionally, we explored the immune cell infiltration and tumor mutational burden (TMB) related to these features. Notably, our study proved a novel approach for predicting treatment responses in THCA patients, including those undergoing chemotherapy and targeted therapy.
Collapse
Affiliation(s)
- Pu Xie
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qinglei Yin
- Guangdong Geriatric Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China;
| | - Shu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dalong Song
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| |
Collapse
|
33
|
Liu Z, Yuan H, Wang Y, Li K, Suo C, Jin L, Ding C, Chen X. Proteogenomic Analysis Identifies a Causal Association between Plasma Apolipoprotein B Levels and Liver Cancer Risk. J Proteome Res 2024; 23:4055-4066. [PMID: 39091241 DOI: 10.1021/acs.jproteome.4c00397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Liver oncogenesis is accompanied by discernible protein changes in the bloodstream. By employing plasma proteomic profiling, we can delve into the molecular mechanisms of liver cancer and pinpoint potential biomarkers. In this nested case-control study, we applied liquid chromatography-tandem mass spectrometry for proteome profiling in baseline plasma samples. Differential protein expression was determined and was subjected to functional enrichment, network, and Mendelian randomization (MR) analyses. We identified 193 proteins with notable differential levels between the groups. Of these proteins, MR analysis offered a compelling negative association between apolipoprotein B (APOB) and liver cancer. This association was further corroborated in the UK Biobank cohort: genetically predicted APOB levels were associated with a 31% (95% CI 19-42%) decreased risk of liver cancer; and phenotypic analysis indicated an 11% (95% CI 8-14%) decreased liver cancer risk for every 0.1 g/L increase of circulating APOB levels. Multivariable MR analysis suggested that the hepatic fat content might fully mediate the APOB-liver cancer connection. In summary, we identified some plasma proteins, particularly APOB, as potential biomarkers of liver cancer. Our findings underscore the intricate link between lipid metabolism and liver cancer, offering hints for targeted prophylactic strategies and early detection.
Collapse
Affiliation(s)
- Zhenqiu Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai 200438, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou 225316, China
| | - Huangbo Yuan
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai 200438, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou 225316, China
| | - Yunzhi Wang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Kai Li
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Chen Suo
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai 200032, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai 200438, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou 225316, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| |
Collapse
|
34
|
Chen Y, Deng X, Li Y, Han Y, Peng Y, Wu W, Wang X, Ma J, Hu E, Zhou X, Shen E, Zeng S, Cai C, Qin Y, Shen H. Comprehensive molecular classification predicted microenvironment profiles and therapy response for HCC. Hepatology 2024; 80:536-551. [PMID: 38537130 PMCID: PMC11332383 DOI: 10.1097/hep.0000000000000869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 02/07/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND AND AIMS Tumor microenvironment (TME) heterogeneity leads to a discrepancy in survival prognosis and clinical treatment response for patients with HCC. The clinical applications of documented molecular subtypes are constrained by several issues. APPROACH AND RESULTS We integrated 3 single-cell data sets to describe the TME landscape and identified 6 prognosis-related cell subclusters. Unsupervised clustering of subcluster-specific markers was performed to generate transcriptomic subtypes. The predictive value of these molecular subtypes for prognosis and treatment response was explored in multiple external HCC cohorts and the Xiangya HCC cohort. TME features were estimated using single-cell immune repertoire sequencing, mass cytometry, and multiplex immunofluorescence. The prognosis-related score was constructed based on a machine-learning algorithm. Comprehensive single-cell analysis described TME heterogeneity in HCC. The 5 transcriptomic subtypes possessed different clinical prognoses, stemness characteristics, immune landscapes, and therapeutic responses. Class 1 exhibited an inflamed phenotype with better clinical outcomes, while classes 2 and 4 were characterized by a lack of T-cell infiltration. Classes 5 and 3 indicated an inhibitory tumor immune microenvironment. Analysis of multiple therapeutic cohorts suggested that classes 5 and 3 were sensitive to immune checkpoint blockade and targeted therapy, whereas classes 1 and 2 were more responsive to transcatheter arterial chemoembolization treatment. Class 4 displayed resistance to all conventional HCC therapies. Four potential therapeutic agents and 4 targets were further identified for high prognosis-related score patients with HCC. CONCLUSIONS Our study generated a clinically valid molecular classification to guide precision medicine in patients with HCC.
Collapse
Affiliation(s)
- Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangying Deng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yin Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yinghui Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xinwen Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiayao Ma
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Erya Hu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Edward Shen
- Department of Life Science, McMaster University, Hamilton, Ontario, Canada
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiming Qin
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
35
|
Lin Y, Wang S, Li Z, Zhou Y, Wang R, Wang Y, Chen Y. Short-Term Statin Therapy Induces Hepatic Insulin Resistance Through HNF4α/PAQR9/PPM1α Axis Regulated AKT Phosphorylation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403451. [PMID: 38970167 PMCID: PMC11425881 DOI: 10.1002/advs.202403451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/24/2024] [Indexed: 07/08/2024]
Abstract
Statins, the first-line medication for dyslipidemia, are linked to an increased risk of type 2 diabetes. But exactly how statins cause diabetes is yet unknown. In this study, a developed short-term statin therapy on hyperlipidemia mice show that hepatic insulin resistance is a cause of statin-induced diabetes. Statin medication raises the expression of progesterone and adiponectin receptor 9 (PAQR9) in liver, which inhibits insulin signaling through degradation of protein phosphatase, Mg2+/Mn2+ dependent 1 (PPM1α) to activate ERK pathway. STIP1 homology and U-box containing protein 1 (STUB1) is found to mediate ubiquitination of PPM1α promoted by PAQR9. On the other hand, decreased activity of hepatocyte nuclear factor 4 alpha (HNF4α) seems to be the cause of PAQR9 expression under statin therapy. The interventions on PAQR9, including deletion of PAQR9, caloric restriction and HNF4α activation, are all effective treatments for statin-induced diabetes, while liver specific over-expression of PPM1α is another possible tactic. The results reveal the importance of HNF4α-PAQR9-STUB1-PPM1α axis in controlling the statin-induced hepatic insulin resistance, offering a fresh insight into the molecular mechanisms underlying statin therapy.
Collapse
Affiliation(s)
- Yijun Lin
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361016, China
| | - Shuying Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zixuan Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yuling Zhou
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361016, China
| | - Ruiying Wang
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361016, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361016, China
| | - Yan Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
36
|
Pan J, Li Q, Zhu J. Unveiling EFNB2 as a Key Player in Sorafenib Resistance: Insights from Bioinformatics Analysis and Functional Validation in Hepatocellular Carcinoma. Biochem Genet 2024:10.1007/s10528-024-10903-5. [PMID: 39212854 DOI: 10.1007/s10528-024-10903-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Sorafenib resistance has become a big hurdle for treating advanced HCC; thus, identifying novel targets to overcome sorafenib resistance is of great importance. Thanks to the massive progress in the sequencing and data analysis, high-throughput screening of novel targets in HCC development has been extensively used in recent years. In present study, we harnessed the public dataset and aimed to identify novel targets related to sorafenib resistance in HCC via bioinformatics analysis and in vitro validation. This study examined three GEO datasets (GSE140202, GSE143233, GSE182593) and identified 20 common DEGs. Functional enrichment analysis suggested these DEGs might play a role in regulating drug resistance pathways. PPI network analysis pinpointed 14 hub genes, with EFNB2 showing high connectivity to other genes. Subsequent in vitro experiments demonstrated that EFNB2 was up-regulated in sorafenib-resistant HCC cells. EFNB2 suppression sensitized HepG2 and Huh7 sorafenib-resistant cells. Furthermore, EFNB2 knockdown increased caspase-3/-7 activities and hindered EMT in sorafenib-resistant HCC cells. Conversely, EFNB2 overexpression promoted sorafenib resistance, decreased caspase-3/-7 activity, and enhanced EMT in HCC cells. Overall, this study identified 14 promising genes potentially linked to sorafenib resistance in HCC, with EFNB2 emerging as a potential contributor to this resistance mechanism.
Collapse
Affiliation(s)
- Junli Pan
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Quanxi Li
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Junli Zhu
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang, 222000, China.
| |
Collapse
|
37
|
Venafra V, Sacco F, Perfetto L. SignalingProfiler 2.0 a network-based approach to bridge multi-omics data to phenotypic hallmarks. NPJ Syst Biol Appl 2024; 10:95. [PMID: 39179556 PMCID: PMC11343843 DOI: 10.1038/s41540-024-00417-6] [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: 01/30/2024] [Accepted: 07/31/2024] [Indexed: 08/26/2024] Open
Abstract
Unraveling how cellular signaling is remodeled upon perturbation is crucial for understanding disease mechanisms and identifying potential drug targets. In this pursuit, computational tools generating mechanistic hypotheses from multi-omics data have invaluable potential. Here, we present a newly implemented version (2.0) of SignalingProfiler, a multi-step pipeline to draw mechanistic hypotheses on the signaling events impacting cellular phenotypes. SignalingProfiler 2.0 derives context-specific signaling networks by integrating proteogenomic data with the prior knowledge-causal network. This is a freely accessible and flexible tool that incorporates statistical, footprint-based, and graph algorithms to accelerate the integration and interpretation of multi-omics data. Through a benchmarking process on three proof-of-concept studies, we demonstrate the tool's ability to generate hierarchical mechanistic networks recapitulating novel and known perturbed signaling and phenotypic outcomes, in both human and mice contexts. In summary, SignalingProfiler 2.0 addresses the emergent need to derive biologically relevant information from complex multi-omics data by extracting interpretable networks.
Collapse
Affiliation(s)
- Veronica Venafra
- Ph.D. Program in Cellular and Molecular Biology, Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Francesca Sacco
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy.
| | - Livia Perfetto
- Department of Biology and Biotechnologies 'C.Darwin', University of Rome 'La Sapienza', Rome, Italy.
| |
Collapse
|
38
|
Liu M, Wu E, Pan F, Tian K, Fu J, Yu Y, Guo Z, Ma Y, Wei A, Yu X, Zhan C, Qian J. Effects of drug-induced liver injury on the in vivo fate of liposomes. Eur J Pharm Biopharm 2024; 201:114389. [PMID: 38945407 DOI: 10.1016/j.ejpb.2024.114389] [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: 04/02/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024]
Abstract
Liposomes represent one of the most extensively studied nano-carriers due to their potential in targeted drug delivery. However, the complex in vivo fate, particularly under pathological conditions, presents challenges for clinical translation of liposomal therapeutics. Liver serves as the most important organ for liposome accumulation and metabolism. Unfortunately, the fate of liposomes under pathological liver conditions has been significantly overlooked. This study aimed to investigate the in vivo pharmacokinetic profile and biodistribution profile of liposomes under drug-induced liver injury (DILI) conditions. Two classic DILI animal models, i.e. acetaminophen-induced acute liver injury (AILI) and triptolide-induced subacute liver injury (TILI), were established to observe the effect of pathological liver conditions on the in vivo performance of liposomes. The study revealed significant changes in the in vivo fate of liposomes following DILI, including prolonged blood circulation and enhanced hepatic accumulation of liposomes. Changes in the composition of plasma proteins and mononuclear phagocyte system (MPS)-related cell subpopulations collectively led to the altered in vivo fate of liposomes under liver injury conditions. Despite liver injury, macrophages remained the primary cells responsible for liposomes uptake in liver, with the recruited monocyte-derived macrophages exhibiting enhanced ability to phagocytose liposomes under pathological conditions. These findings indicated that high capture of liposomes by the recruited hepatic macrophages not only offered potential solutions for targeted delivery, but also warned the clinical application of patients under pathological liver conditions.
Collapse
Affiliation(s)
- Mengyuan Liu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai 201203, PR China
| | - Ercan Wu
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China
| | - Feng Pan
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai 201203, PR China
| | - Kaisong Tian
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China
| | - Jiaru Fu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yifei Yu
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China
| | - Zhiwei Guo
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China
| | - Yinyu Ma
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China
| | - Anqi Wei
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China
| | - Xiaoyue Yu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai 201203, PR China.
| | - Changyou Zhan
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China.
| | - Jun Qian
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai 201203, PR China.
| |
Collapse
|
39
|
Greig JA, Martins KM, Breton C, Lamontagne RJ, Zhu Y, He Z, White J, Zhu JX, Chichester JA, Zheng Q, Zhang Z, Bell P, Wang L, Wilson JM. Integrated vector genomes may contribute to long-term expression in primate liver after AAV administration. Nat Biotechnol 2024; 42:1232-1242. [PMID: 37932420 PMCID: PMC11324525 DOI: 10.1038/s41587-023-01974-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/29/2023] [Indexed: 11/08/2023]
Abstract
The development of liver-based adeno-associated virus (AAV) gene therapies is facing concerns about limited efficiency and durability of transgene expression. We evaluated nonhuman primates following intravenous dosing of AAV8 and AAVrh10 vectors for over 2 years to better define the mechanism(s) of transduction that affect performance. High transduction of non-immunogenic transgenes was achieved, although expression declined over the first 90 days to reach a lower but stable steady state. More than 10% of hepatocytes contained single nuclear domains of vector DNA that persisted despite the loss of transgene expression. Greater reductions in vector DNA and RNA were observed with immunogenic transgenes. Genomic integration of vector sequences, including complex concatemeric structures, were detected in 1 out of 100 cells at broadly distributed loci that were not in proximity to genes associated with hepatocellular carcinoma. Our studies suggest that AAV-mediated transgene expression in primate hepatocytes occurs in two phases: high but short-lived expression from episomal genomes, followed by much lower but stable expression, likely from integrated vectors.
Collapse
Affiliation(s)
- Jenny A Greig
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelly M Martins
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Camilo Breton
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - R Jason Lamontagne
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yanqing Zhu
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhenning He
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John White
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jing-Xu Zhu
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica A Chichester
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qi Zheng
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhe Zhang
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter Bell
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lili Wang
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
40
|
He T, Xu B, Ma H. TLK2 promotes progression of hepatocellular carcinoma through Wnt/β-catenin signaling. Transl Cancer Res 2024; 13:3729-3741. [PMID: 39145094 PMCID: PMC11319953 DOI: 10.21037/tcr-23-2264] [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: 12/11/2023] [Accepted: 05/21/2024] [Indexed: 08/16/2024]
Abstract
Background Hepatocellular carcinoma is a widespread cancer worldwide, ranking as the fifth most frequent cancer and the fourth leading cause of cancer-related deaths. According to comprehensive research, TLK2, a phosphorylated kinase, has been discovered to play a crucial role in promoting tumor development. However, the prognostic significance and influence of TLK2 on hepatocellular carcinoma tumor cells and the immune microenvironment remain unexplored, warranting further investigation. The aim of this study was to investigate the role of TLK2 in promoting the development of hepatocellular carcinoma. Methods The present study utilized The Cancer Genome Atlas (TCGA) database and other databases as training sets to examine the expression of TLK2 and its prognostic significance. The findings were subsequently validated through cell proliferation assays and cell colony assays. Gene set enrichment analysis (GSEA) was employed to investigate the tumor-related biological processes associated with TLK2 in hepatocellular carcinoma, while the relationship between TLK2 expression and Wnt/β-catenin signaling pathway was analyzed via TCGA dataset analysis. Western blotting and immunofluorescence assays were used to confirm the experimental results. Results TLK2 showed higher expression levels in tumor tissues than in normal tissues. Alpha fetoprotein (AFP), T stage, pathological stage, and histological grade were significantly associated with TLK2 expression. High TLK2 expression correlated with worse overall survival (OS) [hazard ratio (HR) =1.62, 95% confidence interval (CI): 1.14-2.29, P=0.007], progression-free survival (PFS) (HR =1.88, 95% CI: 1.40-2.52, P<0.001) and disease specific survival (DSS) (HR =1.86, 95% CI: 1.18-2.93, P=0.007) in the training and validation sets. Both univariate and multivariate analyses showed that TLK2 was an independent prognostic factor. GSEA showed that TLK2 was significantly enriched in tumor-related biological processes. TLK2 induced the activation of β-catenin signaling, resulting in sustained tumor growth. Methyl thiazolyl tetrazolium (MTT) and colony formation assays demonstrated that TLK2 could promote hepatocellular carcinoma progression. Furthermore, TLK2 showed a significant association with β-catenin in the Wnt pathway. Conclusions TLK2 represents an independent prognostic factor in hepatocellular carcinoma and can promote cancer progression via the β-catenin signaling pathway.
Collapse
Affiliation(s)
- Ting He
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Borui Xu
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Haiqing Ma
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Department of Oncology, Heyuan Hospital of Guangdong Provincial People’s Hospital, Heyuan People’s Hospital, Heyuan, China
| |
Collapse
|
41
|
Mestareehi A. Global Gene Expression Profiling and Bioinformatics Analysis Reveal Downregulated Biomarkers as Potential Indicators for Hepatocellular Carcinoma. ACS OMEGA 2024; 9:26075-26096. [PMID: 38911766 PMCID: PMC11191119 DOI: 10.1021/acsomega.4c01496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/25/2024]
Abstract
Objective: The study aimed to elucidate the significance of CLEC4G, CAMK2β, SLC22A1, CBFA2T3, and STAB2 in the prognosis of hepatocellular carcinoma (HCC) patients and their associated molecular biological characteristics. Additionally, the research sought to identify new potential biomarkers with therapeutic and diagnostic relevance for clinical applications. Methods and Materials: We utilized a publicly available high throughput phosphoproteomics and proteomics data set of HCC to focus on the analysis of 12 downregulated phosphoproteins in HCC. Our approach integrates bioinformatic analysis with pathway analysis, encompassing gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and the construction of a protein-protein interaction (PPI) network. Results: In total, we quantified 11547 phosphorylation sites associated with 4043 phosphoproteins from a cohort of 159 HCC patients. Within this extensive data set, our specific focus was on 19 phosphorylation sites displaying significant downregulation (log2 FC ≤ -2 with p-values < 0.0001). Remarkably, our investigation revealed distinct pathways exhibiting differential regulation across multiple dimensions, including the genomic, transcriptomic, proteomic, and phosphoproteomic levels. These pathways encompass a wide range of critical cellular processes, including cellular component organization, cell cycle control, signaling pathways, transcriptional and translational control, and metabolism. Furthermore, our bioinformatics analysis unveiled noteworthy insights into the subcellular localizations, biological processes, and molecular functions associated with these proteins and phosphoproteins. Within the context of the PPI network, we identified 12 key genes CLEC4G, STAB2, ADH1A, ADH1B, CAMK2B, ADH4, CHGB, PYGL, ADH1C, AKAP12, CBFA2T3, and SLC22A1 as the top highly interconnected hub genes. Conclusions: The findings related to CLEC4G, ADH1B, SLC22A1, CAMK2β, CBFA2T3, and STAB2 indicate their reduced expression in HCC, which is associated with an unfavorable prognosis. Furthermore, the results of KEGG and GO pathway analyses suggest that these genes may impact liver cancer by engaging various targets and pathways, ultimately promoting the progression of hepatocellular carcinoma. These results underscore the significant potential of CLEC4G, ADH1B, SLC22A1, CAMK2β, CBFA2T3, and STAB2 as key contributors to HCC development and advancement. This insight holds promise for identifying therapeutic targets and charting research avenues to enhance our understanding of the intricate molecular mechanisms underlying hepatocellular carcinoma.
Collapse
Affiliation(s)
- Aktham Mestareehi
- Department
of Pharmaceutical Sciences, Faculty of Pharmacy, Isra University, P.O. Box 22, Amman 11622, Jordan
- School
of Medicine, The Ohio State University, Columbus, Ohio 43202, United States
- Department
of Pharmaceutical Sciences, School of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan 48201, United States
| |
Collapse
|
42
|
Todorović N, Amedei A. Metadherin-driven promotion of cancer stem cell phenotypes and its effect on immunity in hepatocellular carcinoma. World J Gastroenterol 2024; 30:2624-2628. [PMID: 38855151 PMCID: PMC11154677 DOI: 10.3748/wjg.v30.i20.2624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/24/2024] [Accepted: 05/09/2024] [Indexed: 05/27/2024] Open
Abstract
In this editorial we provide commentary on the article published by Wang et al, featured in the recent issue of the World Journal of Gastroenterology in 2024. We focus on the metadherin (MTDH), also known as astrocyte elevated gene-1 or lysine rich CEACAM1, and its effects on cancer stem cells (CSCs) and immunity in hepatocellular carcinoma (HCC). HCC is the most common primary liver cancer and one of the leading causes of cancer-related deaths worldwide. Most HCC cases develop in the context of liver cirrhosis. Among the pivotal mechanisms of carcinogenesis are gene mutations, dysregulation of diverse signaling pathways, epigenetic alterations, hepatitis B virus-induced hepatocarcinogenesis, chronic inflammation, impact of tumor microenvironment, oxidative stress. Over the years, extensive research has been conducted on the MTDH role in various tumor pathologies, such as lung, breast, ovarian, gastric, hepatocellular, colorectal, renal carcinoma, neuroblastoma, melanoma, and leukemias. Specifically, its involvement in tumor development processes including transformation, apoptosis evasion, angiogenesis, invasion, and metastasis via multiple signaling pathways. It has been demonstrated that knockdown or knockout of MTDH disrupt tumor development and metastasis. In addition, numerous reports have been carried out regarding the MTDH influence on HCC, demonstrating its role as a predictor of poor prognosis, aggressive tumor phenotypes prone to metastasis and recurrence, and exhibiting significant potential for therapy resistance. Finally, more studies finely investigated the influence of MTDH on CSCs. The CSCs are a small subpopulation of tumor cells that sharing traits with normal stem cells like self-renewal and differentiation abilities, alongside a high plasticity that alters their phenotype. Beyond their presumed role in tumor initiation, they can drive also disease relapse, metastasis, and resistance to chemo and radiotherapy.
Collapse
Affiliation(s)
- Nevena Todorović
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
- Clinic for Infectious and Tropical Diseases, University Clinical Centre of Serbia, Belgrade 11000, Serbia
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| |
Collapse
|
43
|
Blukacz L, Nuciforo S, Fucile G, Trulsson F, Duthaler U, Wieland S, Heim MH. Inhibition of the transmembrane transporter ABCB1 overcomes resistance to doxorubicin in patient-derived organoid models of HCC. Hepatol Commun 2024; 8:e0437. [PMID: 38696353 PMCID: PMC11068137 DOI: 10.1097/hc9.0000000000000437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/12/2024] [Indexed: 05/04/2024] Open
Abstract
BACKGROUND Transarterial chemoembolization is the first-line treatment for intermediate-stage HCC. However, the response rate to transarterial chemoembolization varies, and the molecular mechanisms underlying variable responses are poorly understood. Patient-derived hepatocellular carcinoma organoids (HCCOs) offer a novel platform to investigate the molecular mechanisms underlying doxorubicin resistance. METHODS We evaluated the effects of hypoxia and doxorubicin on cell viability and cell cycle distribution in 20 patient-derived HCCO lines. The determinants of doxorubicin response were identified by comparing the transcriptomes of sensitive to resistant HCCOs. Candidate genes were validated by pharmacological inhibition. RESULTS Hypoxia reduced the proliferation of HCCOs and increased the number of cells in the G0/G1 phase of the cell cycle, while decreasing the number in the S phase. The IC50s of the doxorubicin response varied widely, from 29nM to >1µM. Doxorubicin and hypoxia did not exhibit synergistic effects but were additive in some HCCOs. Doxorubicin reduced the number of cells in the G0/G1 and S phases and increased the number in the G2 phase under both normoxia and hypoxia. Genes related to drug metabolism and export, most notably ABCB1, were differentially expressed between doxorubicin-resistant and doxorubicin-sensitive HCCOs. Small molecule inhibition of ABCB1 increased intracellular doxorubicin levels and decreased drug tolerance in resistant HCCOs. CONCLUSIONS The inhibitory effects of doxorubicin treatment and hypoxia on HCCO proliferation are variable, suggesting an important role of tumor-cell intrinsic properties in doxorubicin resistance. ABCB1 is a determinant of doxorubicin response in HCCOs. Combination treatment of doxorubicin and ABCB1 inhibition may increase the response rate to transarterial chemoembolization.
Collapse
MESH Headings
- Doxorubicin/pharmacology
- Humans
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- Organoids/drug effects
- Antibiotics, Antineoplastic/pharmacology
- Antibiotics, Antineoplastic/therapeutic use
- Cell Proliferation/drug effects
- Cell Line, Tumor
- Cell Survival/drug effects
- Chemoembolization, Therapeutic
- Cell Cycle/drug effects
Collapse
Affiliation(s)
- Lauriane Blukacz
- Department of Biomedicine, Hepatology Laboratory, University and University Hospital Basel, Basel, Switzerland
| | - Sandro Nuciforo
- Department of Biomedicine, Hepatology Laboratory, University and University Hospital Basel, Basel, Switzerland
| | - Geoffrey Fucile
- sciCORE Center for Scientific Computing and Center for Data Analytics, University of Basel, Basel, Switzerland
| | - Fredrik Trulsson
- Department of Biomedicine, Hepatology Laboratory, University and University Hospital Basel, Basel, Switzerland
| | - Urs Duthaler
- Department of Biomedicine, Clinical Pharmacology and Toxicology, University and University Hospital Basel, Basel, Switzerland
- Department of Pharmaceutical Sciences, Clinical Pharmacology and Toxicology, University of Basel, Basel, Switzerland
| | - Stefan Wieland
- Department of Biomedicine, Hepatology Laboratory, University and University Hospital Basel, Basel, Switzerland
| | - Markus H. Heim
- Department of Biomedicine, Hepatology Laboratory, University and University Hospital Basel, Basel, Switzerland
- University Digestive Health Care Center Basel - Clarunis, Basel, Switzerland
| |
Collapse
|
44
|
Das D, Wang X, Chiu YC, Bouamar H, Sharkey FE, Lopera JE, Lai Z, Weintraub ST, Han X, Zou Y, Chen HIH, Zeballos Torrez CR, Gu X, Cserhati M, Michalek JE, Halff GA, Chen Y, Zheng S, Cigarroa FG, Sun LZ. Integrative multi-omics characterization of hepatocellular carcinoma in Hispanic patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.27.24306447. [PMID: 38746245 PMCID: PMC11092709 DOI: 10.1101/2024.04.27.24306447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background The incidence and mortality rates of hepatocellular carcinoma (HCC) among Hispanics in the United States are much higher than those of non-Hispanic whites. We conducted comprehensive multi-omics analyses to understand molecular alterations in HCC among Hispanic patients. Methods Paired tumor and adjacent non-tumor samples were collected from 31 Hispanic HCC in South Texas (STX-Hispanic) for genomic, transcriptomic, proteomic, and metabolomic profiling. Additionally, serum lipids were profiled in 40 Hispanic and non-Hispanic patients with or without clinically diagnosed HCC. Results Exome sequencing revealed high mutation frequencies of AXIN2 and CTNNB1 in STX Hispanic HCCs, suggesting a predominant activation of the Wnt/β-catenin pathway. The TERT promoter mutation frequency was also remarkably high in the Hispanic cohort. Cell cycles and liver functions were identified as positively- and negatively-enriched, respectively, with gene set enrichment analysis. Gene sets representing specific liver metabolic pathways were associated with dysregulation of corresponding metabolites. Negative enrichment of liver adipogenesis and lipid metabolism corroborated with a significant reduction in most lipids in the serum samples of HCC patients. Two HCC subtypes from our Hispanic cohort were identified and validated with the TCGA liver cancer cohort. The subtype with better overall survival showed higher activity of immune and angiogenesis signatures, and lower activity of liver function-related gene signatures. It also had higher levels of immune checkpoint and immune exhaustion markers. Conclusions Our study revealed some specific molecular features of Hispanic HCC and potential biomarkers for therapeutic management of HCC and provides a unique resource for studying Hispanic HCC.
Collapse
|
45
|
Simonian M, Lu DS, Whitelegge J, Cohn W, Ahuja P, Hsu W, Raman SS. Comparison of LIRADS 5 Image Guided Core Biopsy Derived From Formalin Fixed and Frozen Tissue Cores for Radiogenomics and Radioproteomics Analysis in Well, Moderate and Poorly Differentiated Hepatocellular Carcinoma. GLOBAL MEDICINE AND THERAPEUTICS 2024; 6:https://www.oatext.com/comparison-of-lirads-5-image-guided-core-biopsy-derived-from-formalin-fixed-and-frozen-tissue-cores-for-radiogenomics-and-radioproteomics-analysis-in-well-moderate-and-poorly-differentiated-hepatocellular-carcinoma.php. [PMID: 38818084 PMCID: PMC11138132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
The aim of this pilot study is to evaluate and compare the quality of the genomics and proteomics data obtained from paired Formalin Fixed Paraffin Embedded (FFPE) and frozen (FF) tissue percutaneous core biopsies of Liver Imaging Reporting and Data System 5 (LIRADS 5) hepatocellular carcinoma (HCC) of varying histological grades. The preliminary data identified differentially expressed proteins and genes in poor, moderate and well differentiated HCC biopsies, with a greater efficacy in fresh frozen samples. The data offered valuable insights into the characteristics and suitability of samples for future studies.
Collapse
Affiliation(s)
- Margaret Simonian
- Dept of Radiological Sciences, David Geffen School of Medicine, UCLA, USA
| | - David S.K. Lu
- Dept of Radiological Sciences, David Geffen School of Medicine, UCLA, USA
| | - Julian Whitelegge
- Semel Institute for Neuroscience & Human Behavior, Pasarow mass spectrometry lab, David Geffen School of Medicine, UCLA, USA
| | - Whitaker Cohn
- Semel Institute for Neuroscience & Human Behavior, Pasarow mass spectrometry lab, David Geffen School of Medicine, UCLA, USA
| | - Preeti Ahuja
- Dept of Radiological Sciences, David Geffen School of Medicine, UCLA, USA
| | - William Hsu
- Dept of Radiological Sciences, David Geffen School of Medicine, UCLA, USA
| | - Steven S. Raman
- Dept of Radiological Sciences, David Geffen School of Medicine, UCLA, USA
| |
Collapse
|
46
|
Zhang Y, Yao L, Chung CR, Huang Y, Li S, Zhang W, Pang Y, Lee TY. KinPred-RNA-kinase activity inference and cancer type classification using machine learning on RNA-seq data. iScience 2024; 27:109333. [PMID: 38523792 PMCID: PMC10959666 DOI: 10.1016/j.isci.2024.109333] [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: 08/26/2023] [Revised: 12/07/2023] [Accepted: 02/21/2024] [Indexed: 03/26/2024] Open
Abstract
Kinases as important enzymes can transfer phosphate groups from high-energy and phosphate-donating molecules to specific substrates and play essential roles in various cellular processes. Existing algorithms for kinase activity from phosphorylated proteomics data are often costly, requiring valuable samples. Moreover, methods to extract kinase activities from bulk RNA sequencing data remain undeveloped. In this study, we propose a computational framework KinPred-RNA to derive kinase activities from bulk RNA-sequencing data in cancer samples. KinPred-RNA framework, using the extreme gradient boosting (XGBoost) regression model, outperforms random forest regression, multiple linear regression, and support vector machine regression models in predicting kinase activities from cancer-related RNA sequencing data. Efficient gene signatures from the LINCS-L1000 dataset were used as inputs for KinPred-RNA. The results highlight its potential to be related to biological function. In conclusion, KinPred RNA constitutes a significant advance in cancer research by potentially facilitating the identification of cancer.
Collapse
Affiliation(s)
- Yuntian Zhang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen 518172, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Lantian Yao
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen 518172, China
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Chia-Ru Chung
- Department of Computer Science and Information Engineering, National Central University, Taoyuan 320953, Taiwan
| | - Yixian Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen 518172, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Shangfu Li
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Wenyang Zhang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Yuxuan Pang
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Tzong-Yi Lee
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| |
Collapse
|
47
|
Jiang H, Wang Y, Wen D, Yu R, Esa SS, Lv K, Feng Q, Liu J, Li F, He L, Di X, Zhang S. Targeting C21orf58 is a Novel Treatment Strategy of Hepatocellular Carcinoma by Disrupting the Formation of JAK2/C21orf58/STAT3 Complex. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306623. [PMID: 38342622 PMCID: PMC11022693 DOI: 10.1002/advs.202306623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/22/2024] [Indexed: 02/13/2024]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Functionally uncharacterized genes are an attractive repository to explore candidate oncogenes. It is demonstrated that C21orf58 displays an oncogenic role in promoting cell growth, tumorigenesis and sorafenib resistance of HCC cells by abnormal activation of STAT3 signaling. Mechanistically, a novel manner to regulate STAT3 signaling that adaptor C21orf58 forms a ternary complex is reveal with N-terminal domain of STAT3 and SH2 domain of JAK2, by which C21orf58 overactivates wild-type STAT3 by facilitating its phosphorylation mediated by JAK2, and hyper-activates of constitutively mutated STAT3 due to preferred binding with C21orf58 and JAK2. Moreover, it is validated that inhibition of C21orf58 with drug alminoprofen, selected by virtual screening, could effectively repress the viability and tumorigenesis of HCC cells. Therefore, it is identified that C21orf58 functions as an oncogenic adaptor, reveal a novel regulatory mechanism of JAK2/STAT3 signaling, explain the cause of abnormal activity of activated mutants of STAT3, and explore the attractive therapeutic potential by targeting C21orf58 in HCC.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Biomedical InformaticsSchool of Life SciencesCentral South UniversityChangshaHunan410013P. R. China
| | - Yang Wang
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangshaHunan410013P. R. China
| | - Doudou Wen
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangshaHunan410013P. R. China
| | - Rongji Yu
- Department of Biomedical InformaticsSchool of Life SciencesCentral South UniversityChangshaHunan410013P. R. China
| | - Sayed S Esa
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangshaHunan410013P. R. China
| | - Kefeng Lv
- School of Biomedical ScienceHunan UniversityChangshaHunan410013P. R. China
| | - Qing Feng
- School of Biomedical ScienceHunan UniversityChangshaHunan410013P. R. China
| | - Jing Liu
- Department of Biochemistry and Molecular BiologySchool of Life SciencesCentral South UniversityChangsha410013P. R. China
| | - Faxiang Li
- Center for Medical GeneticsSchool of Life SciencesCentral South UniversityChangsha410013P. R. China
| | - Lan He
- School of Biomedical ScienceHunan UniversityChangshaHunan410013P. R. China
| | - Xiaotang Di
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangshaHunan410013P. R. China
| | - Shubing Zhang
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangshaHunan410013P. R. China
| |
Collapse
|
48
|
Ramirez CFA, Taranto D, Ando-Kuri M, de Groot MHP, Tsouri E, Huang Z, de Groot D, Kluin RJC, Kloosterman DJ, Verheij J, Xu J, Vegna S, Akkari L. Cancer cell genetics shaping of the tumor microenvironment reveals myeloid cell-centric exploitable vulnerabilities in hepatocellular carcinoma. Nat Commun 2024; 15:2581. [PMID: 38519484 PMCID: PMC10959959 DOI: 10.1038/s41467-024-46835-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/12/2024] [Indexed: 03/25/2024] Open
Abstract
Myeloid cells are abundant and plastic immune cell subsets in the liver, to which pro-tumorigenic, inflammatory and immunosuppressive roles have been assigned in the course of tumorigenesis. Yet several aspects underlying their dynamic alterations in hepatocellular carcinoma (HCC) progression remain elusive, including the impact of distinct genetic mutations in shaping a cancer-permissive tumor microenvironment (TME). Here, in newly generated, clinically-relevant somatic female HCC mouse models, we identify cancer genetics' specific and stage-dependent alterations of the liver TME associated with distinct histopathological and malignant HCC features. Mitogen-activated protein kinase (MAPK)-activated, NrasG12D-driven tumors exhibit a mixed phenotype of prominent inflammation and immunosuppression in a T cell-excluded TME. Mechanistically, we report a NrasG12D cancer cell-driven, MEK-ERK1/2-SP1-dependent GM-CSF secretion enabling the accumulation of immunosuppressive and proinflammatory monocyte-derived Ly6Clow cells. GM-CSF blockade curbs the accumulation of these cells, reduces inflammation, induces cancer cell death and prolongs animal survival. Furthermore, GM-CSF neutralization synergizes with a vascular endothelial growth factor (VEGF) inhibitor to restrain HCC outgrowth. These findings underscore the profound alterations of the myeloid TME consequential to MAPK pathway activation intensity and the potential of GM-CSF inhibition as a myeloid-centric therapy tailored to subsets of HCC patients.
Collapse
Affiliation(s)
- Christel F A Ramirez
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daniel Taranto
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Masami Ando-Kuri
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marnix H P de Groot
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Efi Tsouri
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Zhijie Huang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Daniel de Groot
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Roelof J C Kluin
- Genomics Core facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daan J Kloosterman
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joanne Verheij
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jing Xu
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Serena Vegna
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Leila Akkari
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
49
|
Wu TH, Lin TY, Yang PM, Li WT, Yeh CT, Pan TL. Scutellaria baicalensis Induces Cell Apoptosis and Elicits Mesenchymal-Epithelial Transition to Alleviate Metastatic Hepatocellular Carcinoma via Modulating HSP90β. Int J Mol Sci 2024; 25:3073. [PMID: 38474318 DOI: 10.3390/ijms25053073] [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: 01/18/2024] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Hepatocellular carcinoma is one of the most common malignant tumors in the world and shows strong metastatic potential. Current medicine for hepatocellular carcinoma therapy is invalid, while Scutellaria baicalensis Georgi exhibits the pharmaceutical potential to treat liver diseases and liver cancer. Herein, we verified the inhibitory properties and the pivotal molecules regimented by Scutellaria baicalensis on advanced hepatocellular carcinoma. At first, the viability of SK-Hep-1 cells was significantly reduced under treatment of Scutellaria baicalensis extract in a dose-dependent manner without affecting the growth of normal hepatocyte. Scutellaria baicalensis extract application could remarkably cause apoptosis of SK-Hep-1 cells through p53/cytochrome C/poly-ADP ribose polymerase cascades and arrest the cell cycle at the G1/S phase by downregulating cyclin-dependent kinases. Meanwhile, administration of Scutellaria baicalensis extract remarkably attenuated the migration capability as well as suppressed matrix metalloproteinase activity of advanced hepatocellular carcinoma cells. The proteome profiles and network analysis particularly implied that exposure to Scutellaria baicalensis extract downregulated the expression of HSP90β, and the clinical stage of hepatocellular carcinoma is also positively correlated with the HSP90β level. Combined treatment of Scutellaria baicalensis extract and HSP90β siRNAs could markedly enhance the ubiquitination activity and the degradation of vimentin to subsequently inhibit the metastatic property of SK-Hep-1 cells. Moreover, application of Scutellaria baicalensis extract and HSP90β siRNAs depleted phosphorylation of AKT, which stimulated the expression of p53 and consecutively triggered cell apoptosis. These findings suggest that HSP90β may be a prospective target for the effective therapy of advanced hepatocellular carcinoma via accelerating apoptosis of hepatocellular carcinoma cells and eliciting mesenchymal-epithelial transition with the administration of Scutellaria baicalensis extract.
Collapse
Affiliation(s)
- Tung-Ho Wu
- Surgical Critical Care Division of Cardiovascular Surgical Department, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| | - Tung-Yi Lin
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Keelung, Keelung 204, Taiwan
| | - Pei-Ming Yang
- TMU Research Center of Cancer Translational Medicine, Taipei 110, Taiwan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Tai Li
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112, Taiwan
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Tai-Long Pan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Research Center for Food and Cosmetic Safety and Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
| |
Collapse
|
50
|
Gajos-Michniewicz A, Czyz M. WNT/β-catenin signaling in hepatocellular carcinoma: The aberrant activation, pathogenic roles, and therapeutic opportunities. Genes Dis 2024; 11:727-746. [PMID: 37692481 PMCID: PMC10491942 DOI: 10.1016/j.gendis.2023.02.050] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/28/2022] [Accepted: 02/14/2023] [Indexed: 09/12/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a liver cancer, highly heterogeneous both at the histopathological and molecular levels. It arises from hepatocytes as the result of the accumulation of numerous genomic alterations in various signaling pathways, including canonical WNT/β-catenin, AKT/mTOR, MAPK pathways as well as signaling associated with telomere maintenance, p53/cell cycle regulation, epigenetic modifiers, and oxidative stress. The role of WNT/β-catenin signaling in liver homeostasis and regeneration is well established, whereas in development and progression of HCC is extensively studied. Herein, we review recent advances in our understanding of how WNT/β-catenin signaling facilitates the HCC development, acquisition of stemness features, metastasis, and resistance to treatment. We outline genetic and epigenetic alterations that lead to activated WNT/β-catenin signaling in HCC. We discuss the pivotal roles of CTNNB1 mutations, aberrantly expressed non-coding RNAs and complexity of crosstalk between WNT/β-catenin signaling and other signaling pathways as challenging or advantageous aspects of therapy development and molecular stratification of HCC patients for treatment.
Collapse
Affiliation(s)
- Anna Gajos-Michniewicz
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz 92-215, Poland
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz 92-215, Poland
| |
Collapse
|