1
|
Xu Y, Gao Z, Liu J, Yang Q, Xu S. Role of gut microbiome in suppression of cancers. Gut Microbes 2025; 17:2495183. [PMID: 40254597 PMCID: PMC12013426 DOI: 10.1080/19490976.2025.2495183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 03/23/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025] Open
Abstract
The pathogenesis of cancer is closely related to the disruption of homeostasis in the human body. The gut microbiome plays crucial roles in maintaining the homeostasis of its host throughout lifespan. In recent years, a large number of studies have shown that dysbiosis of the gut microbiome is involved in the entire process of cancer initiation, development, and prognosis by influencing the host immune system and metabolism. Some specific intestinal bacteria promote the occurrence and development of cancers under certain conditions. Conversely, some other specific intestinal bacteria suppress the oncogenesis and progression of cancers, including inhibiting the occurrence of cancers, delaying the progression of cancers and boosting the therapeutic effect on cancers. The promoting effects of the gut microbiome on cancers have been comprehensively discussed in the previous review. This article will review the latest advances in the roles and mechanisms of gut microbiome in cancer suppression, providing a new perspective for developing strategies of cancer prevention and treatment.
Collapse
Affiliation(s)
- Yao Xu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Zhaoyu Gao
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Jiaying Liu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Qianqian Yang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Shunjiang Xu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| |
Collapse
|
2
|
Liang Y, Du M, Li X, Gao J, Li Q, Li H, Li J, Gao X, Cong H, Huang Y, Li X, Wang L, Cui J, Gan Y, Tu H. Upregulation of Lactobacillus spp. in gut microbiota as a novel mechanism for environmental eustress-induced anti-pancreatic cancer effects. Gut Microbes 2025; 17:2470372. [PMID: 39988618 PMCID: PMC11853549 DOI: 10.1080/19490976.2025.2470372] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/01/2024] [Accepted: 02/17/2025] [Indexed: 02/25/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with limited effective treatment options. Emerging evidence links enriched environment (EE)-induced eustress to PDAC inhibition. However, the underlying mechanisms remain unclear. In this study, we explored the role of gut microbiota in PDAC-suppressive effects of EE. We demonstrated that depletion of gut microbiota with antibiotics abolished EE-induced tumor suppression, while fecal microbiota transplantation (FMT) from EE mice significantly inhibited tumor growth in both subcutaneous and orthotopic PDAC models housed in standard environment. 16S rRNA sequencing revealed that EE enhanced gut microbiota diversity and selectively enriched probiotic Lactobacillus, particularly L. reuteri. Treatment with L. reuteri significantly suppressed PDAC tumor growth and increased natural killer (NK) cell infiltration into the tumor microenvironment. Depletion of NK cells alleviated the anti-tumor effects of L. reuteri, underscoring the essential role of NK cell-mediated immunity in anti-tumor response. Clinical analysis of PDAC patients showed that higher fecal Lactobacillus abundance correlated with improved progression-free and overall survival, further supporting the therapeutic potential of L. reuteri in PDAC. Overall, this study identifies gut microbiota as a systemic regulator of PDAC under psychological stress. Supplementation of psychobiotic Lactobacillus may offer a novel therapeutic strategy for PDAC.
Collapse
Affiliation(s)
- Yiyi Liang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Du
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Gao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huimin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Gao
- School of Basic Medicine, Fudan University, Shanghai, China
| | - Hui Cong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yimeng Huang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinran Li
- School of Basic Medicine, Fudan University, Shanghai, China
| | - Liwei Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiujie Cui
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Gan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Tu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Pettas T, Lachanoudi S, Karageorgos FF, Ziogas IA, Fylaktou A, Papalois V, Katsanos G, Antoniadis N, Tsoulfas G. Immunotherapy and liver transplantation for hepatocellular carcinoma: Current and future challenges. World J Transplant 2025; 15:98509. [DOI: 10.5500/wjt.v15.i2.98509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/03/2024] [Accepted: 11/07/2024] [Indexed: 02/21/2025] Open
Abstract
Despite existing curative options like surgical removal, tissue destruction techniques, and liver transplantation for early-stage hepatocellular carcinoma (HCC), the rising incidence and mortality rates of this global health burden necessitate continuous exploration of novel therapeutic strategies. This review critically assesses the dynamic treatment panorama for HCC, focusing specifically on the burgeoning role of immunotherapy in two key contexts: early-stage HCC and downstaging advanced HCC to facilitate liver transplant candidacy. It delves into the unique immunobiology of the liver and HCC, highlighting tumor-mediated immune evasion mechanisms. Analyzing the diverse immunotherapeutic approaches including checkpoint inhibitors, cytokine modulators, vaccines, oncolytic viruses, antigen-targeting antibodies, and adoptive cell therapy, this review acknowledges the limitations of current diagnostic markers alpha-fetoprotein and glypican-3 and emphasizes the need for novel biomarkers for patient selection and treatment monitoring. Exploring the rationale for neoadjuvant and adjuvant immunotherapy in early-stage HCC, current research is actively exploring the safety and effectiveness of diverse immunotherapeutic approaches through ongoing clinical trials. The review further explores the potential benefits and challenges of combining immunotherapy and liver transplant, highlighting the need for careful patient selection, meticulous monitoring, and novel strategies to mitigate post-transplant complications. Finally, this review delves into the latest findings from the clinical research landscape and future directions in HCC management, paving the way for optimizing treatment strategies and improving long-term survival rates for patients with this challenging malignancy.
Collapse
Affiliation(s)
- Theodoros Pettas
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, Thessaloniki 54642, Greece
| | - Sofia Lachanoudi
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, Thessaloniki 54642, Greece
| | - Filippos F Karageorgos
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, Thessaloniki 54642, Greece
| | - Ioannis A Ziogas
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Asimina Fylaktou
- Department of Immunology, National Peripheral Histocompatibility Center, Hippokration General Hospital, Thessaloniki 54642, Greece
| | - Vassilios Papalois
- Department of Transplant Surgery, Imperial College Renal and Transplant Centre, London W12 0HS, United Kingdom
| | - Georgios Katsanos
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University of Thessaloniki, School of Medicine, Thessaloniki 54642, Greece
| | - Nikolaos Antoniadis
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, Thessaloniki 54642, Greece
| | - Georgios Tsoulfas
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University of Thessaloniki, School of Medicine, Thessaloniki 54642, Greece
| |
Collapse
|
4
|
Bai JS, Zou LK, Liu YY, Zhong LH, Chen J, Chen JX, Zhao BQ, Liu RC, Sun BT, Zhou B. Classical swine fever virus utilizes stearoyl-CoA desaturase 1-mediated lipid metabolism to facilitate viral replication. J Virol 2025; 99:e0055125. [PMID: 40387378 DOI: 10.1128/jvi.00551-25] [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: 03/30/2025] [Accepted: 04/28/2025] [Indexed: 05/20/2025] Open
Abstract
Viral infections can significantly alter cellular lipid metabolism by modulating key rate-limiting enzymes, including fatty acid synthase (FASN), stearoyl-CoA desaturase 1 (SCD1), and acetyl-CoA carboxylase (ACC). Our previous study revealed the pivotal role of FASN in lipid droplet (LD) synthesis and the promotion of classical swine fever virus (CSFV) replication. However, the roles of the other two key enzymes in CSFV infection remain unexplored. In this study, we screened a library of 96 lipid metabolism-targeted compounds and identified an antiviral inhibitor of SCD1, a rate-limiting enzyme in monounsaturated fatty acid synthesis, that inhibits CSFV replication. Suppressing SCD1 activity through inhibitors or small interfering RNA knockdown reduces CSFV proliferation. However, this suppression is reversed by adding SCD1 active products (oleic acid/palmitoleic acid [OA/PA]), highlighting the essential role of SCD1 in CSFV proliferation. Mechanistically, CSFV non-structural protein p7 interacts with SCD1 and recruits it to the viral replication complex (VRC) during infection. Importantly, CSFV infection activates the endoplasmic reticulum stress pathway IRE1α/XBP1, which positively regulates SCD1 expression, leading to increased production of triglyceride (TG) and LDs and subsequently enhancing CSFV replication. In summary, our study elucidates the critical role of SCD1 in the CSFV life cycle and highlights its potential as an antiviral target for developing new therapies against Flaviviridae.IMPORTANCEUnderstanding the virus's pathogenesis within the host is essential for advancing antiviral therapeutics and vaccine development. Previous studies have demonstrated that classical swine fever virus (CSFV) leverages host lipid metabolic rate-limiting enzymes, such as fatty acid synthase (FASN), to support viral replication. This study identified stearoyl-CoA desaturase 1 (SCD1), a key enzyme in monounsaturated fatty acid biosynthesis, as a novel regulator of CSFV replication. Mechanistically, the viral non-structural protein p7 mediates the recruitment of SCD1 to the endoplasmic reticulum (ER), facilitating the formation of viral replication complexes (VRCs). Additionally, our findings showed that viral infection activated the ER stress pathway IRE1α/XBP1, which upregulated SCD1 expression and promoted the synthesis of triglycerides (TG) and lipid droplets (LDs). This study provides insights into the metabolic reprogramming triggered by viral infection to support replication and underscores the intricate crosstalk between ER stress and lipid metabolism during CSFV infection. These findings have significant implications for identifying novel antiviral targets against CSFV.
Collapse
Affiliation(s)
- Ji-Shan Bai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Lin-Ke Zou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Ya-Yun Liu
- Institute of Animal Husbandry and Veterinary Science, Livestock and Poultry Epidemic Diseases Research Center of Anhui Province, Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Lin-Han Zhong
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jing Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Jin-Xia Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Bing-Qian Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Rong-Chao Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Bo-Tao Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Bin Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
5
|
Wachsmuth LP, Srinivasan ES, Puviindran BJ, Haskell-Mendoza AP, DeSpenza T, Fecci PE. Autonomic modulation of the immune response and implications for CNS malignancies. NPJ Precis Oncol 2025; 9:168. [PMID: 40483275 PMCID: PMC12145445 DOI: 10.1038/s41698-025-00957-y] [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: 10/08/2024] [Accepted: 05/21/2025] [Indexed: 06/11/2025] Open
Abstract
While the central nervous system (CNS) has long been known to regulate global physiologic processes, its role in regulating immune responses has only relatively recently been appreciated. Specifically, CNS input via the autonomic nervous system (ANS) is increasingly emerging as a crucial modulator of immune responses in numerous pathologies, though understanding of the role of these pathways in malignancy is limited. Herein, we provide an overview of CNS-immune signaling pathways, outline the evidence of ANS inputs to immune organs, provide a detailed description of the impact of ANS signaling on immune cell functions, and consider the implications of ANS-immune regulation for the antitumor immune response and CNS inflammation, with a specific focus on how these factors coalesce to impact the antitumor immune response in intracranial malignancies. This review concludes by highlighting the need to better understand cancer neuro-immunology, the tripartite interactions of malignancy and immune cells within the unique niche of the nervous system.
Collapse
Affiliation(s)
- Lucas P Wachsmuth
- Brain Tumor Immunotherapy Program, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
- Medical Science Training Program, Duke University, Durham, NC, USA
| | - Ethan S Srinivasan
- Brain Tumor Immunotherapy Program, Duke University, Durham, NC, USA
- School of Medicine, Duke University, Durham, NC, USA
| | - Bhairavy J Puviindran
- Brain Tumor Immunotherapy Program, Duke University, Durham, NC, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Aden P Haskell-Mendoza
- Brain Tumor Immunotherapy Program, Duke University, Durham, NC, USA
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Tyrone DeSpenza
- Brain Tumor Immunotherapy Program, Duke University, Durham, NC, USA
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Peter E Fecci
- Brain Tumor Immunotherapy Program, Duke University, Durham, NC, USA.
- Department of Pathology, Duke University, Durham, NC, USA.
- School of Medicine, Duke University, Durham, NC, USA.
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Neurosurgery, Duke University, Durham, NC, USA.
- Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC, USA.
| |
Collapse
|
6
|
Baldi S, Alnaggar M, AL-Mogahed M, Khalil KAA, Zhan X. Monoclonal antibody immune therapy response instrument for stratification and cost-effective personalized approaches in 3PM-guided pan cancer management. EPMA J 2025; 16:465-503. [PMID: 40438490 PMCID: PMC12106254 DOI: 10.1007/s13167-025-00403-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/06/2025] [Indexed: 06/01/2025]
Abstract
Background Immune checkpoint inhibitors (ICIs), such as anti-PD-1, anti-PD-L1, and anti-CTLA-4 therapies, have revolutionized cancer treatment by harnessing the body's immune system to eliminate cancer cells. Despite their considerable promise, the efficacy of ICIs significantly differs based on tumor types and specific patient conditions, highlighting the necessity for personalized approaches in the framework of predictive preventive personalized medicine (PPPM; 3PM). Main body This review proposes a stratification instrument within the 3PM framework to enhance the therapeutic efficacy of ICIs across Pan-cancer. Predictive approaches need to be utilized to enhance the effectiveness of ICIs. For example, biomarkers such as particular genetic alterations and metabolic pathways provide key information on patient treatment responses. To predict treatment outcomes, uncover resistance mechanisms, and tailor medications, we examine biomarkers including PDL-1 and CTLA4. Focusing on cancers like melanoma, bladder, and renal cell carcinoma, we highlight advances in combination therapies and cellular approaches to overcome resistance. We conducted an analysis of clinical trials and public datasets (TCGA, GEO) to evaluate ICI responses across number of cancer types. Survival analysis employed Kaplan-Meier curves and Cox regression. Pan-cancer analysis shows response rates ranging from 19.8% in bladder cancer to > 39% in melanoma when combination therapy is used, emphasizing the potential of 3PM to improve outcomes. By exploring resistance mechanisms and emerging therapeutic innovations, we propose a cost-effective model for better patient stratification and care. Validation of this model requires standardized biomarkers and prospective trials, promising a shift toward precision oncology. Conclusion Within the 3PM framework, this review addresses the urgent need for cost-effective stratification tools and adaptive combinatorial strategies to optimize outcomes.
Collapse
Affiliation(s)
- Salem Baldi
- Department of Medical Laboratory Diagnostics, School of Medical Technology, Shaoyang University, Shaoyang, 422000 China
- Department of Medical Laboratory Diagnostics, Al-Thawra General Hospital, Al Hudaydah, Yemen
| | - Mohammed Alnaggar
- Department of Oncology, South Hubei Cancer Hospital, Chibi, Xianning, 437000 Hubei China
| | - Maged AL-Mogahed
- Department of Urology, The First Bethune Hospital of Jilin University, Changchun, 130012 China
| | - Khalil A. A. Khalil
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 61922 Bisha, Saudi Arabia
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
7
|
Liang G, Ma Y, Deng P, Li S, He C, He H, Liu H, Fan Y, Li Z. Role of cell-based therapies in digestive disorders: Obstacles and opportunities. Regen Ther 2025; 29:1-18. [PMID: 40124469 PMCID: PMC11925584 DOI: 10.1016/j.reth.2025.02.009] [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: 12/22/2024] [Revised: 02/01/2025] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
Stem cell-based therapies have emerged as a promising frontier in the treatment of gastrointestinal disorders, offering potential solutions for challenges posed by conventional treatments. This review comprehensively examines recent advancements in cell-based therapeutic strategies, particularly focusing on stem cell applications, immunotherapy, and cellular therapies for digestive diseases. It highlights the successful differentiation of enteric neural progenitors from pluripotent stem cells and their application in animal models, such as Hirschsprung disease. Furthermore, the review evaluates clinical trials and experimental studies demonstrating the potential of stem cells in regenerating damaged tissues, modulating immune responses, and promoting healing in conditions like Crohn's disease and liver failure. By addressing challenges, such as scalability, immunogenicity, and ethical considerations, the review underscores the translational opportunities and obstacles in realizing the clinical potential of these therapies. Concluding with an emphasis on future directions, the study provides insights into optimizing therapeutic efficacy and fostering innovations in personalized medicine for digestive disorders.
Collapse
Affiliation(s)
- Guodong Liang
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Yuehan Ma
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Ping Deng
- Medical Department, Jilin Cancer Hospital, Changchun 130012, China
| | - Shufeng Li
- First Department of Gynecological Tumor, Jilin Cancer Hospital, Changchun 130012, China
| | - Chunyan He
- Department of Anaesthesia, Jilin Cancer Hospital, Changchun 130012, China
| | - Haihang He
- Department of Otorhinolaryngology, Oral Maxillofacial, Head and Neck, Jilin Cancer Hospital, Changchun 130012, China
| | - Hairui Liu
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Yunda Fan
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Ze Li
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| |
Collapse
|
8
|
Zhang L, Yang Y, Li Y, Wang C, Bian C, Wang H, Wang F. Epigenetic regulation of histone modifications in glioblastoma: recent advances and therapeutic insights. Biomark Res 2025; 13:80. [PMID: 40450300 DOI: 10.1186/s40364-025-00788-w] [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: 03/13/2025] [Accepted: 05/14/2025] [Indexed: 06/03/2025] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor, characterized by its aggressive behavior, limited treatment options, and poor prognosis. Despite advances in surgery, radiotherapy, and chemotherapy, the median survival of GBM patients remains disappointingly short. Recent studies have underscored the critical role of histone modifications in GBM malignant progression and therapy resistance. Histones, protein components of chromatin, undergo various modifications, including acetylation and methylation. These modifications significantly affect gene expression, thereby promoting tumorigenesis and resistance to therapy. Targeting histone modifications has emerged as a promising therapeutic approach. Numerous pre-clinical studies have evaluated histone modification agents in GBM, including histone deacetylase inhibitors and histone methyltransferase inhibitors. These studies demonstrate that modulating histone modifications can alter gene expression patterns, inhibit tumor growth, induce apoptosis, and sensitize tumor cells to conventional treatments. Some agents have advanced to clinical trials, aiming to translate preclinical efficacy into clinical benefit. However, clinical outcomes remain suboptimal, as many agents fail to significantly improve GBM patient prognosis. These challenges are attributed to the complexity of histone modification networks and the adaptive responses of the tumor microenvironment. This review provides a comprehensive overview of epigenetic regulation mechanisms involving histone modifications in GBM, covering their roles in tumor development, tumor microenvironment remodeling, and therapeutic resistance. Additionally, the review discusses current clinical trials targeting histone modifications in GBM, highlighting successes, limitations, and future perspectives.
Collapse
Affiliation(s)
- Li Zhang
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Yang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanchu Li
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chenyu Wang
- Yuexiu District, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan 2 Road, Guangzhou City, Guangdong Province, China
| | - Chenbin Bian
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongbin Wang
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Wang
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
9
|
Kounatidis D, Vallianou NG, Karampela I, Grivakou E, Dalamaga M. The intricate role of adipokines in cancer-related signaling and the tumor microenvironment: Insights for future research. Semin Cancer Biol 2025; 113:130-150. [PMID: 40412490 DOI: 10.1016/j.semcancer.2025.05.013] [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: 03/19/2025] [Revised: 05/05/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Obesity represents a global health challenge, with adipose tissue acting as a highly active endocrine organ that synthesizes and secretes a diverse array of bioactive proteins, known as adipokines. These cell signaling molecules regulate metabolic equilibrium, inflammatory cascades, and immune surveillance, exerting substantial systemic effects. A growing body of evidence has also highlighted their key role in cancer biology, through their intricate impact on oncogenic signaling networks and the tumor microenvironment (TME). The TME, a highly dynamic and heterotypic network composed of malignant cells, infiltrating immune cells, stromal constituents, and extracellular matrix elements, facilitates tumor evolution and immune evasion. Among adipokines, adiponectin and leptin have been extensively studied. Research has shown that adiponectin exhibits tumor-suppressive properties, whereas leptin enhances proliferative, angiogenic, and inflammatory pathways that promote malignancy. However, these effects are context-dependent and, at times, contradictory across different studies. Furthermore, the functional landscape of adipokines in cancer extends beyond these paradigms, with emerging research identifying a broader spectrum of novel adipokines involved in cancer reprogramming. This review delineates the molecular interplay between adipokines and oncogenic pathways, elucidating their mechanistic contributions to TME crosstalk and immune modulation. Additionally, we examine their potential as diagnostic and prognostic biomarkers and assess their viability as therapeutic targets for precision oncology. By integrating current evidence and identifying unresolved questions, this review aims to refine our understanding of adipokine-driven tumor biology and establish a platform for future research.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens 11527, Greece.
| | - Natalia G Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, Athens 15126, Greece.
| | - Irene Karampela
- Second Department of Critical Care, Medical School, Attikon General University Hospital, University of Athens, Athens 12461, Greece.
| | - Evgenia Grivakou
- Emergency Department, Limassol General Hospital, Limassol 4131, Cyprus.
| | - Maria Dalamaga
- Department of Biological Chemistry, National and Kapodistrian University of Athens, Athens 11527, Greece.
| |
Collapse
|
10
|
Gebauer N, Wang SS. The Role of EBV in the Pathogenesis of Diffuse Large B-Cell Lymphoma. Curr Top Microbiol Immunol 2025. [PMID: 40399571 DOI: 10.1007/82_2025_296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
There are multiple established risk factors for DLBCL; these risk factors share an underlying biology, which generally cause immune dysfunction, spanning immunosuppression to chronic inflammation. EBV is an established risk factor for DLBCL and approximately 10% of DLBCLs are EBV-positive. EBV is a ubiquitous infection, and it is thus among populations that are immunocompromised, by age or medically defined, where EBV-positive DLBCLs arise. In this chapter, we review the current classification, epidemiology, clinical, pathology, and molecular characteristics of EBV-positive DLBCL, and discuss the role of EBV in lymphoma tumorigenesis. We further discuss current and novel treatments aimed at the NFκB pathway and other targets.
Collapse
Affiliation(s)
- Niklas Gebauer
- Klinik für Hämatologie und Onkologie, UKSH Campus Lübeck, Lübeck, Germany
| | - Sophia S Wang
- Division of Computational and Quantitative Medicine, City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
11
|
Plewa P, Kiełbowski K, Mentel O, Figiel K, Bakinowska E, Becht R, Banach B, Pawlik A. Bacteria and Carcinogenesis and the Management of Cancer: A Narrative Review. Pathogens 2025; 14:509. [PMID: 40430828 PMCID: PMC12114594 DOI: 10.3390/pathogens14050509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/17/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
There is a widely known relationship between certain microbes and cancer progression. For instance, Helicobacter pylori is associated with the occurrence of gastric cancer, while HPV is associated with cervical and head and neck cancers. Recent studies have uncovered novel and important associations between bacterial presence and tumor formation and treatment response. Apart from the influence of the intestinal microbiome on cancer, the local activity of bacteria affects disease properties as well. Bacteria can localize within tumors in less vascularized niches. Their presence mediates the activity of signaling pathways, which contribute to tumorigenesis. Furthermore, they affect the composition of the tumor microenvironment, a highly complex structure composed of immunoregulatory cells and secreted inflammatory mediators. Recently, researchers have analyzed the properties of bacteria to develop novel anticancer strategies. The aim of this review is to discuss the latest findings regarding the relationships between bacteria and cancer and the properties of bacteria that could be used to kill cancer cells.
Collapse
Affiliation(s)
- Paulina Plewa
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.P.)
| | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.P.)
- Department of Clinical Oncology, Chemotherapy and Cancer Immunotherapy, Pomeranian Medical University, 71-252 Szczecin, Poland
| | - Oliwia Mentel
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.P.)
| | - Karolina Figiel
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.P.)
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.P.)
| | - Rafał Becht
- Department of Clinical Oncology, Chemotherapy and Cancer Immunotherapy, Pomeranian Medical University, 71-252 Szczecin, Poland
| | - Bolesław Banach
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.P.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.P.)
| |
Collapse
|
12
|
Ma T, Guo WW, Zhang M, He W, Dongzhi C, Gongye X, Xia P, Chai Y, Chen Z, Zhu Y, Qu C, Liu J, Yang Z, Ma W, Tian M, Yuan Y. Tumor-derived exosomal CCT6A serves as a matchmaker introducing chemokines to tumor-associated macrophages in pancreatic ductal adenocarcinoma. Cell Death Dis 2025; 16:382. [PMID: 40374617 DOI: 10.1038/s41419-025-07720-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 04/16/2025] [Accepted: 05/06/2025] [Indexed: 05/17/2025]
Abstract
M2-polarized tumor-associated macrophages (TAMs) are a key factor contributing to the poor prognosis of pancreatic ductal adenocarcinoma (PDAC). While various factors within the tumor microenvironment (TME) drive their formation, the role of PDAC-derived exosomes in this process remains unclear. We aim to clarify the regulatory impacts of tumor-derived exosomes to TAMs. After the intratumoral injection to subcutaneous tumor of C57BL/6 mice, we demonstrated PDAC-derived exosomes exacerbate PDAC progression, accompanied with upregulated M2 phenotype of TAMs and unaffected proliferation signatures. Through intratumoral injection model and multi-Omics analyses, we identified CCT6A as a novel tumor-derived exosomal protein, bridging TAMs M2 polarization and PDAC prognosis. Co-culture with exosomes derived from CCT6Ahigh PDAC leads to greater M2 phenotype of TAMs via PI3K-AKT signaling. According to proteomics data, chemokines' abundance reduces over tenfold once exosomal CCT6A absence, including CXCL1, CXCL3, CCL20 and CCL5, whose interaction with CCT6A in PDAC cells was confirmed by interactomics data. Moreover, we found silencing CCT6A abrogated the antagonism effects of CD47 antibody immunotherapy. Our findings implied that the subunit of the T-complex protein Ring Complex (TRiC) CCT6A serves as a matchmaker during exosome-mediated chemokines transfer from PDAC to TAMs. Silencing CCT6A effectively sensitized PDAC to CD47 antibody immunotherapy in vivo.
Collapse
Affiliation(s)
- Tianyin Ma
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
| | - Wing-Wa Guo
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
| | - Minghe Zhang
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
| | - Wenzhi He
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
| | - Cairang Dongzhi
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
| | - Xiangdong Gongye
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
- Department of Chemistry and Molecular Biology, Sahlgrenska Akademin, Göteborg Universitet, Gothenburg, Vastra Gotalands, Sweden
| | - Peng Xia
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Yibo Chai
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
| | - Zhang Chen
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
| | - Yimin Zhu
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
| | - Chengming Qu
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
| | - Jie Liu
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China
| | - Zhiyong Yang
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China.
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China.
| | - Weijie Ma
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China.
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China.
| | - Ming Tian
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China.
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China.
| | - Yufeng Yuan
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, PR China.
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, PR China.
- Taikang Center for Life and Medical Sciences of Wuhan University, Hubei, PR China.
| |
Collapse
|
13
|
Zhang X, Zhao J, Ge R, Zhang X, Sun H, Guo Y, Wang Y, Chen L, Li S, Yang J, Sun D. Arg-Gly-Asp engineered mesenchymal stem cells as targeted nanotherapeutics against kidney fibrosis by modulating m6A. Acta Biomater 2025; 198:85-101. [PMID: 40158765 DOI: 10.1016/j.actbio.2025.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/17/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Background The recent surge in research on extracellular vesicles has generated considerable interest in their clinical applications. Extracellular vesicles derived from mesenchymal stem cells (MSC-EV) have emerged as a promising cell-free therapy for chronic kidney disease (CKD), offering an alternative to traditional Mesenchymal stem/stromal cells (MSCs) in extracellular vesicle-based nanotherapeutics. However, challenges such as in vivo off-target effects and limited bioavailability have impeded the wider adoption of MSC-EV in clinical settings. Methods Arginyl-glycyl-aspartic acid peptide-modified MSC-EV (RGD-MSC-EV) were developed using a donor cell-assisted membrane modification strategy. The targeting capability and therapeutic efficacy of RGD-MSC-EV were thoroughly evaluated both in vitro and in vivo. Additionally, the mechanisms of RNA N6-methyladenosine (m6A) methylation-mediated angiogenesis were extensively investigated to elucidate how RGD-MSC-EV mitigates renal fibrosis. Results RGD-MSC-EV demonstrated exceptional targeted delivery efficiency, exhibiting optimal biodistribution and retention within the target tissue. This breakthrough positions them as significantly enhanced anti-fibrotic therapeutics. Notably, RGD-MSC-EV sustains the viability of renal peritubular capillary (PTCs) endothelial cells by transporting microRNA-126-5p (miR-126-5p) and modulating alkB homolog 5 (ALKBH5)-mediated m6A modification of SIRT1(Sirtuin 1), a crucial regulator in angiogenesis. By revitalizing endothelial cells and promoting microcirculation, this approach restored oxygen metabolism homeostasis, ultimately delaying fibrogenesis associated with CKD. Conclusions RGD-MSC-EV offers a feasible and effective strategy to alleviate renal interstitial fibrosis by restoring m6A and mitigating the loss of renal PTCs. STATEMENT OF SIGNIFICANCE: Chronic kidney disease (CKD) often leads to renal fibrosis, which worsens disease progression. This study introduces a novel strategy using engineered extracellular vesicles (EVs) derived from mesenchymal stem cells (MSC-EV). By modifying these EVs with RGD peptides, we significantly enhance their targeting ability to hypoxic kidney tissues. The research reveals how these EVs deliver microRNA (miR-126-5p) to restore key molecular mechanisms, stabilizing SIRT1 expression through m6A RNA modifications. This approach promotes blood vessel health and delays fibrosis. Compared to current treatments, RGD-MSC-EV offers a safe, effective, and cell-free therapeutic alternative. These findings advance the understanding of EV-based therapies and their clinical potential, bridging basic research and real-world CKD treatment applications.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Jiaqi Zhao
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Rui Ge
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Xiangyu Zhang
- Department of Nephrology, Ningbo City first Hospital, Ningbo, China
| | - Haihan Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China
| | - Yuhan Guo
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Yanping Wang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Lu Chen
- Department of Rheumatology, Ningbo Medical Treatment Center Li Huili Hospital, Ningbo, China
| | - Shulin Li
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Jing Yang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China
| | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
14
|
Yan B, Fritsche AK, Haußner E, Inamdar TV, Laumen H, Boettcher M, Gericke M, Michl P, Rosendahl J. From Genes to Environment: Elucidating Pancreatic Carcinogenesis Through Genetically Engineered and Risk Factor-Integrated Mouse Models. Cancers (Basel) 2025; 17:1676. [PMID: 40427173 PMCID: PMC12110317 DOI: 10.3390/cancers17101676] [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: 03/28/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic cancer is characterized by late diagnosis, therapy resistance, and poor prognosis, necessitating the exploration of early carcinogenesis and prevention methods. Preclinical mouse models have evolved from cell line-based to human tumor tissue- or organoid-derived xenografts, now to humanized mouse models and genetically engineered mouse models (GEMMs). GEMMs, primarily driven by oncogenic Kras mutations and tumor suppressor gene alterations, offer a realistic platform for investigating pancreatic cancer initiation, progression, and metastasis. The incorporation of inducible somatic mutations and CRISPR-Cas9 screening methods has expanded their utility. To better recapitulate tumor initiation triggered by inflammatory cues, common pancreatic risk factors are being integrated into model designs. This approach aims to decipher the role of environmental factors as secondary or parallel triggers of tumor initiation alongside oncogenic burdens. Emerging models exploring pancreatitis, obesity, diabetes, and other risk factors offer significant translational potential. This review describes current mouse models for studying pancreatic carcinogenesis, their combination with inflammatory factors, and their utility in evaluating pathogenesis, providing guidance for selecting the most suitable models for pancreatic cancer research.
Collapse
Affiliation(s)
- Bin Yan
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Anne-Kristin Fritsche
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany;
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany;
| | - Erik Haußner
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.H.); (M.B.)
| | - Tanvi Vikrant Inamdar
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| | - Helmut Laumen
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| | - Michael Boettcher
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.H.); (M.B.)
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany;
| | - Patrick Michl
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| |
Collapse
|
15
|
Harrington K, Shah K. Harmonizing the Gut Microbiome and Cellular Immunotherapies: The Next Leap in Cancer Treatment. Cells 2025; 14:708. [PMID: 40422211 DOI: 10.3390/cells14100708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 05/06/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
The gut microbiome, a diverse community of microorganisms, plays a key role in shaping the host's immune system and modulating cancer therapies. Emerging evidence highlights its critical influence on the efficacy and toxicity of cell-based immunotherapies, including chimeric antigen receptor T cell, natural killer cell, and stem cell therapies. This review explores the interplay between gut microbiota and cellular immunotherapies, focusing on mechanisms by which microbial metabolites and microbial composition impact treatment outcomes. Furthermore, we discuss strategies to leverage the gut microbiome to optimize therapeutic efficacy and minimize adverse effects. A deeper understanding of the relationship between the gut microbiome and cellular immunotherapies can pave the way for more effective cell-based therapies for cancer.
Collapse
Affiliation(s)
- Kendall Harrington
- Center for Stem Cell and Translational Immunotherapy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Khalid Shah
- Center for Stem Cell and Translational Immunotherapy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
16
|
Liu G, Liu J, Li S, Zhang Y, He R. Exosome-Mediated Chemoresistance in Cancers: Mechanisms, Therapeutic Implications, and Future Directions. Biomolecules 2025; 15:685. [PMID: 40427578 PMCID: PMC12108986 DOI: 10.3390/biom15050685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Chemotherapy resistance represents a formidable obstacle in oncological therapeutics, substantially compromising the efficacy of adjuvant chemotherapy regimens and contributing to unfavorable clinical prognoses. Emerging evidence has elucidated the pivotal involvement of exosomes in the dissemination of chemoresistance phenotypes among tumor cells and within the tumor microenvironment. This review delineates two distinct intra-tumoral resistance mechanisms orchestrated by exosomes: (1) the exosome-mediated sequestration of chemotherapeutic agents coupled with enhanced drug efflux in neoplastic cells, and (2) the horizontal transfer of chemoresistance to drug-sensitive cells through the delivery of bioactive molecular cargo, thereby facilitating the propagation of resistance phenotypes across the tumor population. Furthermore, the review covers current in vivo experimental data focusing on targeted interventions against specific genetic elements and exosomal secretion pathways, demonstrating their potential in mitigating chemotherapy resistance. Additionally, the therapeutic potential of inhibiting exosome-mediated transporter transfer strategy is particularly examined as a promising strategy to overcome tumor resistance mechanisms.
Collapse
Affiliation(s)
| | | | | | - Yumiao Zhang
- School of Chemical Engineering and Technology, School of Synthetic Biology and Biomanufacturing, Frontiers Science Center for Synthetic Biology (Ministry of Education) and State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300350, China; (G.L.); (J.L.); (S.L.)
| | - Ren He
- School of Chemical Engineering and Technology, School of Synthetic Biology and Biomanufacturing, Frontiers Science Center for Synthetic Biology (Ministry of Education) and State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300350, China; (G.L.); (J.L.); (S.L.)
| |
Collapse
|
17
|
Osaki M, Terakura S, Hirano S, Iwasa T, Hatanaka KC, Hatanaka Y, Sunagawa M, Kokuryo T, Adachi Y, Takeuchi Y, Hanajiri R, Sakanaka C, Murata M, Ebata T, Kiyoi H. Development and optimization of Eva1 ( MPZL2) targeting chimeric antigen receptor T cells. J Immunother Cancer 2025; 13:e009825. [PMID: 40341026 DOI: 10.1136/jitc-2024-009825] [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] [Accepted: 04/16/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Whereas chimeric antigen receptor gene modified T (CAR-T) cell therapy has been clinically applied to malignant lymphomas and multiple myeloma, CAR-T cell therapy for solid tumors has so far not reached clinical application. Epithelial V-like antigen 1 (Eva1), transcribed from myelin protein zero-like 2 (MPZL2), is a small surface protein highly expressed on various tumor cells. We selected Eva1 as a novel solid tumor-target antigen because of its broad expression across various tumor types. The purpose of the present study is to develop and optimize CAR-T cells targeting Eva1. METHOD We prepared various humanized single chain variable fragment sequences based on a mouse anti-human Eva1 monoclonal antibody. We constructed six humanized Eva1CAR-Ts and selected one that maintained specificity and good cellular proliferation after antigen stimulation. We further optimized the length of the extracellular spacer domain and the choice of the intracellular domain in vitro and in two different xenograft mouse models. RESULTS We confirmed Eva1 expression on various tumor cell lines by flow cytometry and analysis of public database, but we also observed that normal monocytes weakly expressed Eva1. A combination of short spacer domain and 4-1BB or CD79A/CD40 intracellular domain provided higher treatment efficacy both in vitro and in vivo. The cytokine release on autologous monocyte stimulation to Eva1CAR-T cells was comparable to that on autologous B cell stimulation to CD19CAR-T cells. Humanized Eva1CAR-T cells demonstrated excellent therapeutic efficacy by infusing a single dose of Eva1CAR-T cells (1×106) in both NCI-H1975 lung cancer and CFPAC-1 pancreatic cancer cell line grafted model. CONCLUSIONS In summary, these data suggest that humanized Eva1CAR-T has promising therapeutic potential for the treatment of various Eva1-positive solid tumors. Regarding on-target/off-tumor recognition, further detailed analyses of the Eva1CAR-T cell responses to normal tissues are needed.
Collapse
Affiliation(s)
- Masahide Osaki
- Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seitaro Terakura
- Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiho Hirano
- Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Kanako C Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Yutaka Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Masaki Sunagawa
- Division of Surgical Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshio Kokuryo
- Division of Surgical Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshitaka Adachi
- Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Takeuchi
- Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryo Hanajiri
- Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Makoto Murata
- Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoki Ebata
- Division of Surgical Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hitoshi Kiyoi
- Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
18
|
Han Y, Cao B, Tang J, Wang J. A comprehensive multi-omics analysis uncovers the associations between gut microbiota and pancreatic cancer. Front Microbiol 2025; 16:1592549. [PMID: 40376462 PMCID: PMC12078283 DOI: 10.3389/fmicb.2025.1592549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/17/2025] [Indexed: 05/18/2025] Open
Abstract
Pancreatic cancer is one of the most lethal malignant neoplasms. Pancreatic cancer is related to gut microbiota, but the associations between its treatment and microbial abundance as well as genetic variations remain unclear. In this study, we collected fecal samples from 58 pancreatic cancer patients including 43 pancreatic ductal adenocarcinoma (PDAC) and 15 non-PDAC, and 40 healthy controls, and shotgun metagenomic sequencing and untargeted metabolome analysis were conducted. PDAC patients were divided into five groups according to treatment and tumor location, including treatment-naive (UT), chemotherapy (CT), surgery combined with chemotherapy (SCT), Head, and body/tail (Tail) groups. Multivariate association analysis revealed that both CT and SCT were associated with increased abundance of Lactobacillus gasseri and Streptococcus equinus. The microbial single nucleotide polymorphisms (SNPs) densities of Streptococcus salivarius, Streptococcus vestibularis and Streptococcus thermophilus were positively associated with CT, while Lachnospiraceae bacterium 2_1_58FAA was positively associated with Head group. Compared with Tail group, the Head group showed positive associations with opportunistic pathogens, such as Escherichia coli, Shigella sonnei and Shigella flexneri. We assembled 424 medium-quality non-redundant metagenome-assembled genomes (nrMAGs) and 276 high-quality nrMAGs. In CT group, indole-3-acetic acid, capsaicin, sinigrin, chenodeoxycholic acid, and glycerol-3-phosphate were increased, and the accuracy of the model based on fecal metabolites reached 0.77 in distinguishing healthy controls and patients. This study identifies the associations between pancreatic cancer treatment and gut microbiota as well as its metabolites, reveals bacterial SNPs are related to tumor location, and extends our knowledge of gut microbiota and pancreatic cancer.
Collapse
Affiliation(s)
- Yang Han
- Medical Innovation Research Division, Chinese PLA General Hospital, Beijing, China
| | - Biyang Cao
- The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jiayue Tang
- The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jing Wang
- The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
19
|
da Silva H, Juniastuti, Amin M, Soares J, Soares M, Malik H, Ximenes A, Bela M, Fernandes B. Genotypes, subtypes, and genetic variability of hepatitis B virus from blood donors in Timor-Leste. Arch Virol 2025; 170:119. [PMID: 40310552 DOI: 10.1007/s00705-025-06305-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/17/2025] [Indexed: 05/02/2025]
Abstract
Timor-Leste experiences high hepatitis B endemicity; however, information about hepatitis B virus (HBV) variants in Timor-Leste is still limited. In this study, we determined genotypes and subtypes and identified mutations in the surface (S), polymerase (P), basal core promoter (BCP), precore (PC), and core (C) genes of HBV isolates from blood donors in Timor-Leste. Sera were examined using serological tests and PCR sequencing. Out of 127 sera tested, 38 (30%) were positive for the hepatitis B S antigen (HBsAg). Thirty-eight sequences of the S and P genes, 22 sequences of the BCP and PC regions, and 23 sequences of C genes were determined and analyzed. The most common genotype/subtype was C/adrq+, followed by B/ayw1, B/adw2, and C/adw2. Several mutations in the S protein that are associated with vaccine escape were identified in samples of genotype C (I110L, S113T, T126I, T143S, Y161F) and B (K122R), some of which might have been from vaccinated individuals. None of the healthy carriers had taken anti-HBV drugs, but one was infected with a virus with a mutation in the P gene associated with anti-HBV drug resistance (Y141F). The mutations A1762T and G1764A in BCP were detected in 18.1-22.7% of the samples. In the PC region, the mutation C1858T was the most frequent, followed by G1896A and G1899A. In the C gene, 13 mutations (P5T, T67N, E77Q, P79Q/A, E83D, V91T, I97L/F, L116I, and P130I/P/T) associated with severe liver disease were identified. Viruses obtained from four healthy carriers who were later found to have died of hepatocellular carcinoma also showed those mutations. In conclusion, among blood donors in Timor-Leste, HBV genotype/subtype C/adrq+ and several mutations in the S and C genes were prevalent. Routine implementation of a national immunization program and monitoring of disease progression in healthy carriers should be considered.
Collapse
Affiliation(s)
- Hendriketa da Silva
- Postdoctoral Fellowship Program, Universitas Airlangga, Surabaya, Indonesia
- Postgraduation and Research Program, Faculty of Medicine and Health Sciences, Universidade Nacional Timor-Loro sae, Dili, Timor-Leste
| | - Juniastuti
- Department of Medical Microbiology, School of Medicine, Universitas Airlangga, Jl. Mayjen. Prof. Dr. Moestopo 47, Surabaya, East Java, 60131, Indonesia.
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia.
| | - Mochamad Amin
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | | | - Miguel Soares
- Hospital Nacional Guido Valadares, Dili, Timor-Leste
| | - Hitler Malik
- Hospital Nacional Guido Valadares, Dili, Timor-Leste
| | | | - Maria Bela
- Hospital Nacional Guido Valadares, Dili, Timor-Leste
| | | |
Collapse
|
20
|
Pele KG, Calderón-Villalba A, Amaveda H, Mora M, Zhang-Zhou J, Pérez MÁ, García-Aznar JM, Alamán-Díez P, García-Gareta E. Novel hydrogel-based cancer-on-a-chip models for growth of 3D multi-cellular structures and investigation of early angiogenesis in pancreatic ductal adenocarcinoma. Colloids Surf B Biointerfaces 2025; 253:114736. [PMID: 40315572 DOI: 10.1016/j.colsurfb.2025.114736] [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: 10/07/2024] [Revised: 03/27/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
Cancer-on-a-chip models have enormous potential for the study of tumour development events. Here, we investigated hydrogels of egg white (EW) and gelatin for growth of 3D multi-cellular structures and investigation of early angiogenesis inside microfluidic devices. We focused on pancreatic ductal adenocarcinoma (PDAC), a devastating gastrointestinal malignancy. EW/gelatin hydrogels were stiffer and showed porous globular structures compared to the fibrous network of collagen I molecules. PANC-1 cells preferentially formed significantly larger spheroids in collagen I than in EW/gelatin hydrogels, whilst cell aggregates in the shape of grape-like clusters were significantly larger and more abundant in EW/gelatin. Cells inside the aggregates showed active cell unions, secreted matrix, and formed active unions with the surrounding EW/gelatin hydrogel. Early stages of PDAC were recreated by co-culture of two different microenvironments, one for PANC-1 and another one for fibroblasts, for investigating the secretion of soluble angiogenic factors, which depended on the role of each factor in the angiogenic and tumorigenic processes. Overall, cancer cell proliferation and establishment of a tumour vasculature were favoured. This study demonstrates the importance of the microenvironment in tumour cells behaviour as well as the complex interplay between the different cells present in PDAC to establish a tumoural vasculature.
Collapse
Affiliation(s)
- Karinna Georgiana Pele
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Alejandro Calderón-Villalba
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Hippolyte Amaveda
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC and University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Mario Mora
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC and University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Jack Zhang-Zhou
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - María Ángeles Pérez
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain
| | - José Manuel García-Aznar
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain
| | - Pilar Alamán-Díez
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Elena García-Gareta
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain; Division of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, London NW3 2QG, United Kingdom.
| |
Collapse
|
21
|
Burge KY, Georgescu C, Zhong H, Wilson AP, Gunasekaran A, Yu Z, Franca A, Eckert JV, Wren JD, Chaaban H. Spatial transcriptomics delineates potential differences in intestinal phenotypes of cardiac and classical necrotizing enterocolitis. iScience 2025; 28:112166. [PMID: 40201118 PMCID: PMC11978348 DOI: 10.1016/j.isci.2025.112166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/20/2024] [Accepted: 03/03/2025] [Indexed: 04/10/2025] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating neonatal gastrointestinal disease, often resulting in multi-organ failure and death. While classical NEC is strictly associated with prematurity, cardiac NEC is a subset of the disease occurring in infants with comorbid congenital heart disease. Despite similar symptomatology, the NEC subtypes vary slightly in presentation and may represent etiologically distinct diseases. We compared ileal spatial transcriptomes of patients with cardiac and classical NEC. Epithelial and immune cells cluster well by cell-type segment and NEC subtype. Differences in metabolism and immune cell activation functionally differentiate the cell-type makeup of the NEC subtypes. The classical NEC phenotype is defined by dysbiosis-induced inflammatory signaling and metabolic acidosis, while that of cardiac NEC involves reduced angiogenesis and endoplasmic reticulum stress-induced apoptosis. Despite subtype-associated clinical and demographic variability, spatial transcriptomics has substantiated pathway and network differences within immune and epithelial segments between cardiac and classical NEC.
Collapse
Affiliation(s)
- Kathryn Y. Burge
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Constantin Georgescu
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Hua Zhong
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Adam P. Wilson
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Aarthi Gunasekaran
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zhongxin Yu
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Addison Franca
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jeffrey V. Eckert
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jonathan D. Wren
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Hala Chaaban
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
22
|
Younis OM, Dhaydel AS, Alghwyeen WF, Abu Hantash NR, Allan LM, Qasem IM, Saeed A. The role of ANGPTL4 in cancer: A meta-analysis of observational studies and multi-omics investigation. PLoS One 2025; 20:e0320343. [PMID: 40233044 PMCID: PMC11999138 DOI: 10.1371/journal.pone.0320343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/16/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Angiopoietin-like protein 4 (ANGPTL4) plays a crucial role in processes such as angiogenesis, inflammation, and metabolism. Despite numerous studies suggesting its involvement in cancer, a definitive role remains unclear. We introduce the first comprehensive meta-analysis and pan-cancer bioinformatics study on ANGPTL4, aiming to unravel its implications across various cancer types. METHODS Moderate-to high-quality observational studies were retrieved from PubMed, Scopus, and Embase. A meta-analysis was conducted using the R package "meta." Survival analysis was performed using GEPIA2 and TIMER2.0. Immune infiltration, mutational burden, and drug resistance analyses was done via GSCAlite. Co-expression and gene set enrichment analyses (GSEA) were carried out using cBioportal and enrichr, respectively. RESULTS Increased ANGPTL4 expression was linked to worse tumor grade (OR = 1.51, P = 0.023), stage (OR = 2.42, P < 0.001), lymph node metastasis (OR = 1.76, P = 0.012), vascular invasion (OR = 2.16, P = 0.01), and lymphatic invasion (OR = 2.20, P < 0.001). Furthermore, ANGPTL4 expression was linked to worse OS (HR = 1.40, 95% CI: 1.29,1.50, P = 0.0001). Single gene level analysis revealed that ANGPTL4 upregulated epithelial-to-mesenchymal transition (EMT) in 23 different cancers. Immune infiltration varied between cancer types, but increased infiltration of cancer-associated fibroblasts was observed in most cancers. Mutation analysis revealed increased alterations in TP53 and CDKN2A in cohorts with ANGPTL4 alterations. GSEA of co-expressed genes revealed involvement in hypoxia, EMT, VEGF-A complex, TGF-B pathways, and extracellular matrix organization. CONCLUSIONS ANGPTL4 plays a significant role in tumor progression via its positive regulation of EMT and angiogenesis, while possibly harboring a TGF-B dependent role in systemic metastasis. Therefore, ANGPTL4 is a suitable target for future drug development.
Collapse
Affiliation(s)
- Osama M. Younis
- Division of Hematology & Oncology, Department of Medicine, University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, United States of America
| | | | | | | | - Leen M. Allan
- School of Medicine, The Hashemite University, Al Zarqaa, Jordan
| | | | - Anwaar Saeed
- Division of Hematology & Oncology, Department of Medicine, University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
23
|
Lv X, Liu J, Islam K, Ruan J, He C, Chen P, Huang C, Wang H, Dhar A, Moness M, Shi D, Murphy S, Zhao X, Yang S, Montoute I, Polakkattil A, Chung A, Ruiz E, Carbajal B, Padavala A, Chen L, Hua G, Chen X, Davis JS, Wang C. Hyperactivated YAP1 is essential for sustainable progression of renal clear cell carcinoma. Oncogene 2025:10.1038/s41388-025-03354-8. [PMID: 40210757 DOI: 10.1038/s41388-025-03354-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 02/12/2025] [Accepted: 03/17/2025] [Indexed: 04/12/2025]
Abstract
The most notable progress in renal clear cell carcinoma (ccRCC) in the past decades is the introduction of drugs targeting the VHL-HIF signaling pathway-associated angiogenesis. However, mechanisms underlying the development of VHL mutation-independent ccRCC are unclear. Here we provide evidence that the disrupted Hippo-YAP signaling contributes to the development of ccRCC independent of VHL alteration. We found that YAP1 and its primary target genes are frequently upregulated in ccRCC and the upregulation of these genes is associated with unfavorable patient outcomes. Research results derived from our in vitro and in vivo experimental models demonstrated that, under normoxic conditions, hyperactivated YAP1 drives the expression of FGFs to stimulate the proliferation of tumor and tumor-associated endothelial cells in an autocrine/paracrine manner. When rapidly growing cancer cells create a hypoxic environment, hyperactivated YAP1 in cancer cells induces the production of VEGF, which promotes the angiogenesis of tumor-associated endothelial cells, leading to improved tumor microenvironment and continuous tumor growth. Our study indicates that hyperactivated YAP1 is essential for maintaining ccRCC progression, and targeting the dual role of hyperactivated YAP1 represents a novel strategy to improve renal carcinoma therapy.
Collapse
Affiliation(s)
- Xiangmin Lv
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiyuan Liu
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kazi Islam
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jinpeng Ruan
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Chunbo He
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peichao Chen
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cong Huang
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hongbo Wang
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anjali Dhar
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Chemistry, Dartmouth College, Hanover, NH, USA
| | - Madelyn Moness
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Davie Shi
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Savannah Murphy
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xingeng Zhao
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Siyi Yang
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Isabelle Montoute
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Aneeta Polakkattil
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andie Chung
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily Ruiz
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Brianna Carbajal
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Stem cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Alekhya Padavala
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Li Chen
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Guohua Hua
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xingcheng Chen
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - John S Davis
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Cheng Wang
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Ding Z, Wang L, Sun J, Zheng L, Tang Y, Tang H. Hepatocellular carcinoma: pathogenesis, molecular mechanisms, and treatment advances. Front Oncol 2025; 15:1526206. [PMID: 40265012 PMCID: PMC12011620 DOI: 10.3389/fonc.2025.1526206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Hepatocellular Carcinoma (HCC), a highly prevalent malignancy, poses a significant global health challenge. Its pathogenesis is intricate and multifactorial, involving a complex interplay of environmental and genetic factors. Viral hepatitis, excessive alcohol consumption, and cirrhosis are known to significantly elevate the risk of developing HCC. The underlying biological processes driving HCC are equally complex, encompassing aberrant activation of molecular signaling pathways, dysregulation of hepatocellular differentiation and angiogenesis, and immune dysfunction. This review delves into the multifaceted nature of HCC, exploring its etiology and the intricate molecular signaling pathways involved in its development. We examine the role of immune dysregulation in HCC progression and discuss the potential of emerging therapeutic strategies, including immune-targeted therapy and tumor-associated macrophage interventions. Additionally, we explore the potential of traditional Chinese medicine (TCM) monomers in inhibiting tumor growth. By elucidating the complex interplay of factors contributing to HCC, this review aims to provide a comprehensive understanding of the disease and highlight promising avenues for future research and therapeutic development.
Collapse
Affiliation(s)
- Zhixian Ding
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Lusheng Wang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Jiting Sun
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Lijie Zheng
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Yu Tang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Heng Tang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| |
Collapse
|
25
|
Tian C, Ye C, Guo H, Lu K, Yang J, Wang X, Ge X, Yu C, Lu J, Jiang L, Zhang Q, Song C. Liver elastography-based risk score for predicting hepatocellular carcinoma risk. J Natl Cancer Inst 2025; 117:761-771. [PMID: 39576686 DOI: 10.1093/jnci/djae304] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 04/08/2025] Open
Abstract
BACKGROUND Liver stiffness measurement (LSM) via vibration-controlled transient elastography accurately assesses fibrosis. We aimed to develop a universal risk score for predicting hepatocellular carcinoma (HCC) development in patients with chronic hepatitis. METHODS We systematically selected predictors and developed the risk prediction model (HCC-LSM) in the hepatitis B virus (HBV) training cohort (n = 2251, median follow-up of 3.2 years). The HCC-LSM model was validated in an independent HBV validation cohort (n = 1191, median follow-up of 5.7 years) and a non-viral chronic liver disease (CLD) extrapolation cohort (n = 1189, median follow-up of 3.3 years). An HCC risk score was then constructed based on a nomogram. An online risk evaluation tool Liver Elastography-Based Hepatocellular Carcinoma Risk Score (LEBER) was developed using ChatGPT4.0. RESULTS Eight routinely available predictors were identified, with LSM levels showing a significant dose-response relationship with HCC incidence (P < .001 by log-rank test). The HCC-LSM model exhibited excellent predictive performance in the HBV training cohort (C-index = 0.866) and the HBV validation cohort (C-index = 0.852), with good performance in the extrapolation CLD cohort (C-index = 0.769). The model demonstrated significantly superior discrimination compared to 6 previous models across the 3 cohorts. Cut-off values of 87.2 and 121.1 for the HCC-LSM score categorized participants into low-, medium-, and high-risk groups. An online public risk evaluation tool (LEBER; http://ccra.njmu.edu.cn/LEBER669.html) was developed to facilitate the use of HCC-LSM. CONCLUSION The accessible, reliable risk score based on LSM accurately predicted HCC development in patients with chronic hepatitis, providing an effective risk assessment tool for HCC surveillance strategies.
Collapse
Affiliation(s)
- Chan Tian
- Health Management Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, P. R. China
| | - Chunyan Ye
- Department of Liver Diseases, The Third People's Hospital of Changzhou, Changzhou 213000, Jiangsu, China
| | - Haiyan Guo
- Health Management Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, P. R. China
| | - Kun Lu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, P. R. China
| | - Juan Yang
- Health Management Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, P. R. China
| | - Xiao Wang
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xinyuan Ge
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, P. R. China
| | - Chengxiao Yu
- Health Management Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, P. R. China
| | - Jing Lu
- Health Management Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, P. R. China
| | - Longfeng Jiang
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qun Zhang
- Health Management Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ci Song
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, P. R. China
| |
Collapse
|
26
|
Saadh MJ, Hussain QM, Alazzawi TS, Fahdil AA, Athab ZH, Yarmukhamedov B, Al-Nuaimi AMA, Alsaikhan F, Farhood B. MicroRNA as Key Players in Hepatocellular Carcinoma: Insights into Their Role in Metastasis. Biochem Genet 2025; 63:1014-1062. [PMID: 39103713 DOI: 10.1007/s10528-024-10897-0] [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: 06/13/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
Liver cancer or hepatocellular carcinoma (HCC) remains the most common cancer in global epidemiology. Both the frequency and fatality of this malignancy have shown an upward trend over recent decades. Liver cancer is a significant concern due to its propensity for both intrahepatic and extrahepatic metastasis. Liver cancer metastasis is a multifaceted process characterized by cell detachment from the bulk tumor, modulation of cellular motility and invasiveness, enhanced proliferation, avoidance of the immune system, and spread either via lymphatic or blood vessels. MicroRNAs (miRNAs) are small non-coding ribonucleic acids (RNAs) playing a crucial function in the intricate mechanisms of tumor metastasis. A number of miRNAs can either increase or reduce metastasis via several mechanisms, such as control of motility, proliferation, attack by the immune system, cancer stem cell properties, altering the microenvironment, and the epithelial-mesenchymal transition (EMT). Besides, two other types of non-coding RNAs, such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) can competitively bind to endogenous miRNAs. This competition results in the impaired ability of the miRNAs to inhibit the expression of the specific messenger RNAs (mRNAs) that are targeted. Increasing evidence has shown that the regulatory axis comprising circRNA/lncRNA-miRNA-mRNA is correlated with the regulation of HCC metastasis. This review seeks to present a thorough summary of recent research on miRNAs in HCC, and their roles in the cellular processes of EMT, invasion and migration, as well as the metastasis of malignant cells. Finally, we discuss the function of the lncRNA/circRNA-miRNA-mRNA network as a crucial modulator of carcinogenesis and the regulation of signaling pathways or genes that are relevant to the metastasis of HCC. These findings have the potential to offer valuable insight into the discovery of novel therapeutic approaches for management of liver cancer metastasis.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | - Tuqa S Alazzawi
- College of Dentist, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Ali A Fahdil
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Bekhzod Yarmukhamedov
- Department of Public Health and Healthcare management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
| | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
27
|
Wang Z, Xu C, Wang Q, Wang Y. Repurposing of nervous system drugs for cancer treatment: recent advances, challenges, and future perspectives. Discov Oncol 2025; 16:396. [PMID: 40133751 PMCID: PMC11936871 DOI: 10.1007/s12672-025-02067-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
The nervous system plays a critical role in developmental biology and oncology, influencing processes from ontogeny to the complex dynamics of cancer progression. Interactions between the nervous system and cancer significantly affect oncogenesis, tumor growth, invasion, metastasis, treatment resistance, inflammation that promotes tumors, and the immune response. A comprehensive understanding of the signal transduction pathways involved in cancer biology is essential for devising effective anti-cancer strategies and overcoming resistance to existing therapies. Recent advances in cancer neuroscience promise to establish a new cornerstone of cancer therapy. Repurposing drugs originally developed for modulating nerve signal transduction represent a promising approach to target oncogenic signaling pathways in cancer treatment. This review endeavors to investigate the potential of repurposing neurological drugs, which target neurotransmitters and neural pathways, for oncological applications. In this context, it aims to bridge the interdisciplinary gap between neurology, psychiatry, internal medicine, and oncology. By leveraging already approved drugs, researchers can utilize existing extensive safety and efficacy data, thereby reducing both the time and financial resources necessary for the development of new cancer therapies. This strategy not only promises to enhance patient outcomes but also to expand the array of available treatments, thereby enriching the therapeutic landscape in oncology.
Collapse
Affiliation(s)
- Zixun Wang
- Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China
| | - Chen Xu
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Qi Wang
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Yudong Wang
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
28
|
Pinto E, Lazzarini E, Pelizzaro F, Gambato M, Santarelli L, Potente S, Zanaga P, Zappitelli T, Cardin R, Burra P, Farinati F, Romualdi C, Boscarino D, Tosello V, Indraccolo S, Russo FP. Somatic Copy Number Alterations in Circulating Cell-Free DNA as a Prognostic Biomarker for Hepatocellular Carcinoma: Insights from a Proof-of-Concept Study. Cancers (Basel) 2025; 17:1115. [PMID: 40227625 PMCID: PMC11988118 DOI: 10.3390/cancers17071115] [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: 02/20/2025] [Revised: 03/13/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Despite advances in hepatocellular carcinoma (HCC) management, prognosis remains poor. Advanced-stage diagnosis often excludes curative treatments, and current biomarkers (e.g., alpha-fetoprotein [AFP]) have limited utility in early detection. Liquid biopsy has emerged as a promising cancer detection tool, with circulating cell-free DNA (ccfDNA) showing significant diagnostic potential. This proof-of-concept study aimed to investigate the potential role of tumor fraction (TF) within ccfDNA as a biomarker in HCC patients. METHODS A total of sixty patients were recruited, including thirteen with chronic liver disease (CLD), twenty-four with cirrhosis, and twenty-three with HCC. Plasma samples were collected, and ccfDNA was extracted for shallow whole genome sequencing (sWGS) analysis. The TF was calculated by focusing on somatic copy number alterations (SCNAs) within the ccfDNA. RESULTS Among patients with CLD and cirrhosis (n = 37), ctDNA was undetectable in all but one cirrhotic patient who exhibited a significant tumor fraction (TF) of 17% and subsequently developed HCC. Conversely, five out of twenty-three HCC patients (21.7%) displayed detectable ctDNA with TF levels ranging from 3.0% to 32.6%. Patients with detectable ctDNA were characterized by more aggressive oncological features, including a higher number of nodules (p = 0.005), advanced-stage disease (60% BCLC C, p = 0.010), and poorer response to therapy (80% PD, p = 0.001). Moreover, the overall survival (OS) was significantly reduced in patients with detectable ctDNA (median OS: 17 months; CI 95% 4.5-26.5) compared to those without (median OS: 24.0 months; CI 95% 7.0-66.0; log-rank p = 0.002). CONCLUSIONS Our results suggest that the analysis of TF by sWGS is a promising non-invasive tool for the identification of HCC with aggressive clinical behavior, whereas it is not sensitive enough for early HCC detection. This molecular assay can improve prognostic stratification in HCC patients.
Collapse
Affiliation(s)
- Elisa Pinto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Elisabetta Lazzarini
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35121 Padua, Italy; (E.L.); (L.S.); (V.T.)
| | - Filippo Pelizzaro
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Martina Gambato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Laura Santarelli
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35121 Padua, Italy; (E.L.); (L.S.); (V.T.)
| | - Sara Potente
- Department of Biology, University of Padova, 35121 Padua, Italy; (S.P.); (C.R.)
| | - Paola Zanaga
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Teresa Zappitelli
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Romilda Cardin
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Patrizia Burra
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Fabio Farinati
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Chiara Romualdi
- Department of Biology, University of Padova, 35121 Padua, Italy; (S.P.); (C.R.)
| | | | - Valeria Tosello
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35121 Padua, Italy; (E.L.); (L.S.); (V.T.)
| | - Stefano Indraccolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35121 Padua, Italy; (E.L.); (L.S.); (V.T.)
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| |
Collapse
|
29
|
Rong Y, Wang M, Ma Y, Liang Y, Ye L, Guo L, Lu R, Wang Y. Serum hepatitis B core antibody as the prognostic factor for diffuse large B-cell lymphoma. Microbiol Spectr 2025; 13:e0317024. [PMID: 40130869 PMCID: PMC12053999 DOI: 10.1128/spectrum.03170-24] [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: 12/04/2024] [Accepted: 02/10/2025] [Indexed: 03/26/2025] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin lymphoma (NHL), strongly associated with viral infections. Although the link between hepatitis B virus (HBV) infection and DLBCL is well-documented, effective clinical markers reflecting HBV-associated DLBCL remain scarce. This study aims to identify prognostic indicators for HBV-associated DLBCL through retrospective analysis of the relationship among tissue marker molecules, HBV serum markers, and clinical prognosis in DLBCL patients. Here, we found the results that DLBCL patients who tested positive for hepatitis B core antibody (HBcAb) had significantly reduced overall survival (OS) rates compared with those who tested negative. Additionally, a strong correlation was observed between an elevated HBcAb-positive rate and reduced expression of the CD23 molecule in DLBCL tissue samples. Stratifying DLBCL patients based on combined HBcAb-CD23 status revealed significant disparities in OS rates. Therefore, integrating CD23 with HBcAb could be applied to prognostic assessments for individuals with HBV-associated DLBCL. This study identifies novel indicators and diagnostic strategies for HBV-associated DLBCL.IMPORTANCEThis study identifies hepatitis B core antibody (HBcAb) as a significant prognostic indicator for hepatitis B virus (HBV)-associated diffuse large B-cell lymphoma (DLBCL). The findings reveal that patients with DLBCL with positive HBcAb have significantly reduced overall survival rates. Additionally, a strong negative correlation is observed between serum HBcAb and the expression of the CD23 molecule in DLBCL tissues. These results highlight the potential of integrating HBcAb and CD23 as prognostic markers in clinical assessments of HBV-associated DLBCL, offering new insights for risk stratification and treatment planning in this patient population.
Collapse
Affiliation(s)
- Yi Rong
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ming Wang
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yaqiong Ma
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yuanchen Liang
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lvyin Ye
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lin Guo
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Renquan Lu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yanchun Wang
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Chen D, Yu R, Cai Y, Zhang H, Jiang Y, Wu Y, Peng XE. Additive interaction between hepatitis B virus infection and tobacco smoking on the risk of gastric cancer in a Chinese population. Infect Agent Cancer 2025; 20:19. [PMID: 40114245 PMCID: PMC11927330 DOI: 10.1186/s13027-025-00648-0] [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: 01/17/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE Although hepatitis B virus (HBV) infection was regarded as a risk factor for liver cancer, the association of HBV infection with gastric cancer (GC) is unclear. In this study, we aim to assess the association of HBV infection with the risk of GC and explore the interaction between HBV infection and other risk factors. METHODS A case-control study was conducted and 409 GC cases and 1275 healthy controls were enrolled in Fujian province, China. Serum hepatitis B surface antigen (HBsAg) was measured and epidemiological data were collected. The association between HBV infection and GC risk was analyzed using logistic regression and meta-analysis method was employed to make estimates more conservative. Meanwhile, multiplicative and additive models were used to explore the interaction between HBV infection and other risk factors. RESULTS The prevalence of serum HBsAg positivity was 13.20% among GC cases and 6.20% among controls. Compared to HBsAg-negative subjects, the adjusted odds ratios (OR) for HBsAg positive were 3.30 [95% confidence interval (CI): (1.84-5.91)]. Compared to HBsAg-negative never smokers, the adjusted OR was 2.00 (95%CI: 1.19-3.34) for HBsAg-negative ever smokers,4.27 (95%CI: 1.97-9.26) for HBsAg-positive never smokers, and 4.73 (95%CI: 1.85-12.08) for HBsAg-positive ever smokers. These evidences indicated super-additive [API (95%CI): 0.78 (0.67-0.90), S (95%CI): 5.45 (3.26-9.08)] between HBV infection and tobacco smoking. No interaction between HBV infection and alcohol drinking was found on the risk of GC. CONCLUSIONS HBV infection increased the risk of GC, and tobacco smoking and HBV infection may positively interact in the development of GC.
Collapse
Affiliation(s)
- Danjing Chen
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Rong Yu
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Yongfeng Cai
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - He Zhang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Yijun Jiang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Yunli Wu
- Key Laboratory of Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Ministry of Education, Fujian Medical University, Fuzhou, 350108, People's Republic of China
| | - Xian-E Peng
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China.
- Key Laboratory of Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Ministry of Education, Fujian Medical University, Fuzhou, 350108, People's Republic of China.
- Department of Epidemiology and Health Statistics, Minhou Country, Xuefu North Road 1st, Shangjie Town, Minhou Country, Fuzhou, 350108, Fujian, China.
| |
Collapse
|
31
|
Ahangari G, Norioun H. The dual role of dopamine and serotonin in cancer progression and inflammation: Mechanisms and therapeutic potential. Neuroscience 2025; 569:184-194. [PMID: 39675693 DOI: 10.1016/j.neuroscience.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/20/2024] [Accepted: 12/07/2024] [Indexed: 12/17/2024]
Abstract
This review examines the critical functions of dopamine and serotonin in the regulation of inflammation and cancer, emphasizing their potential as therapeutic targets. Traditionally recognized for their roles in neural communication, these neurotransmitters are now understood to play substantial roles in immune modulation and tumor progression. We conducted a systematic review of studies published between 2013 and 2024, using databases such as PubMed, Google Scholar, and Scopus, to assess dopamine and serotonin synthesis, receptor activity, and involvement in disease pathways. Findings indicate that dopamine, through its D1 and D2 receptors, exerts both pro- and anti-inflammatory effects, influencing tumor growth and immune responses in cancers such as breast and pancreatic. Similarly, serotonin, particularly through receptors HTR2A and HTR2B, has demonstrated dual roles in cancer progression, impacting the growth and metastasis of cancers such as gastric and colorectal. This review also addresses the interaction between dopamine and serotonin signaling pathways, which may collectively alter immune cell function and tumor microenvironment dynamics, suggesting a promising direction for combined therapeutic approaches. By synthesizing current data on dopamine and serotonin pathways, this review aims to inform the development of targeted therapies that modulate immune responses in inflammation-driven cancers. Our findings underscore the potential of neurotransmitter-based interventions as a novel strategy for managing cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Ghasem Ahangari
- Medical Genetics Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Iran.
| | - Hamid Norioun
- Medical Genetics Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Iran.
| |
Collapse
|
32
|
Kindekov I, Beleva E, Kadish M, Ionchev I, Semerdzhieva N. Hairy Cell Leukemia: A Differential Diagnosis of Hepatitis B-Associated Aplastic Anemia and Syphilis. Hematol Rep 2025; 17:13. [PMID: 40126222 PMCID: PMC11932296 DOI: 10.3390/hematolrep17020013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/25/2025] Open
Abstract
Aplastic anemia occurs with an incidence of 2-5: 1 million people worldwide. However, the frequency of newly diagnosed cases of bone marrow aplasia is greater, and some of these patients present to emergency departments initially. Description of Case: We present the case of a middle-aged man with pancytopenia. In this case, aplastic anemia associated with hepatitis B and syphilis was only the initial diagnosis. An indolent hematologic malignancy-hairy cell leukemia-was diagnosed as the real cause of the bone marrow failure in a clinic of hematology. Conclusions: This clinical case allows us to make a conclusion, albeit not definitively, about the contribution of hepatitis B and syphilis to the clinical manifestation of hairy cell leukemia. A detailed and consistent diagnostic plan is also required in patients presenting with pancytopenia. Failure to diagnose a hepatitis B infection in a patient with malignant hematologic disease would lead to fatal therapeutic errors.
Collapse
Affiliation(s)
- I. Kindekov
- Clinic of Hematology, Military Hospital, 1606 Sofia, Bulgaria; (I.K.); (E.B.)
| | - E. Beleva
- Clinic of Hematology, Military Hospital, 1606 Sofia, Bulgaria; (I.K.); (E.B.)
- QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1784 Sofia, Bulgaria
| | - M. Kadish
- Clinic of Internal Medicine, Hepatology and Gastroenterology, University Hospital ‘SOFIAMED’, 1797 Sofia, Bulgaria;
| | - I. Ionchev
- Clinic of Internal Medicine, University Hospital of Emergency Medicine ‘Pirogov’, 1606 Sofia, Bulgaria
| | - N. Semerdzhieva
- Clinic of Internal Medicine, University Hospital of Emergency Medicine ‘Pirogov’, 1606 Sofia, Bulgaria
| |
Collapse
|
33
|
Li S, Zhou Y, Wang H, Qu G, Zhao X, Wang X, Hou R, Guan Z, Liu D, Zheng J, Shi M. Advances in CAR optimization strategies based on CD28. Front Immunol 2025; 16:1548772. [PMID: 40181986 PMCID: PMC11966486 DOI: 10.3389/fimmu.2025.1548772] [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: 12/20/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy, which utilizes genetic engineering techniques to modify T-cells to achieve specific targeting of cancer cells, has made significant breakthroughs in cancer treatment in recent years. All marketed CAR-T products are second-generation CAR-T cells containing co-stimulatory structural domains, and co-stimulatory molecules are critical for CAR-T cell activation and function. Although CD28-based co-stimulatory molecules have demonstrated potent cytotoxicity in the clinical application of CAR-T cells, they still suffer from high post-treatment relapse rates, poor efficacy durability, and accompanying severe adverse reactions. In recent years, researchers have achieved specific results in enhancing the anti-tumor function of CD28 by mutating its signaling motifs, combining the co-stimulatory structural domains, and modifying other CAR components besides co-stimulation. This paper reviewed the characteristics and roles of CD28 in CAR-T cell-mediated anti-tumor signaling and activation. We explored potential strategies to enhance CAR-T cell efficacy and reduce side effects by optimizing CD28 motifs and CAR structures, aiming to provide a theoretical basis for further clinical CAR-T cell therapy development.
Collapse
Affiliation(s)
- Sijin Li
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Yusi Zhou
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Hairong Wang
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Gexi Qu
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Xuan Zhao
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Xu Wang
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Rui Hou
- College of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zhangchun Guan
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Dan Liu
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Ming Shi
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
34
|
Srivastava A, Vinod PK. A single-cell network approach to decode metabolic regulation in gynecologic and breast cancers. NPJ Syst Biol Appl 2025; 11:26. [PMID: 40082472 PMCID: PMC11906788 DOI: 10.1038/s41540-025-00506-0] [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: 09/19/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
Cancer metabolism is characterized by significant heterogeneity, presenting challenges for treatment efficacy and patient outcomes. Understanding this heterogeneity and its regulatory mechanisms at single-cell resolution is crucial for developing personalized therapeutic strategies. In this study, we employed a single-cell network approach to characterize malignant heterogeneity in gynecologic and breast cancers, focusing on the transcriptional regulatory mechanisms driving metabolic alterations. By leveraging single-cell RNA sequencing (scRNA-seq) data, we assessed the metabolic pathway activities and inferred cancer-specific protein-protein interactomes (PPI) and gene regulatory networks (GRNs). We explored the crosstalk between these networks to identify key alterations in metabolic regulation. Clustering cells by metabolic pathways revealed tumor heterogeneity across cancers, highlighting variations in oxidative phosphorylation, glycolysis, cholesterol, fatty acid, hormone, amino acid, and redox metabolism. Our analysis identified metabolic modules associated with these pathways, along with their key transcriptional regulators. These findings provide insights into the complex interplay between metabolic rewiring and transcriptional regulation in gynecologic and breast cancers, paving the way for potential targeted therapeutic strategies in precision oncology. Furthermore, this pipeline for dissecting coregulatory metabolic networks can be broadly applied to decipher metabolic regulation in any disease at single-cell resolution.
Collapse
Affiliation(s)
- Akansha Srivastava
- Centre for Computational Natural Sciences and Bioinformatics, IIIT, Hyderabad, India
| | - P K Vinod
- Centre for Computational Natural Sciences and Bioinformatics, IIIT, Hyderabad, India.
| |
Collapse
|
35
|
Xie Z, Qu X, Zhang J, Huang Y, Runhan Z, Tang D, Li N, Wang Z, Luo X. Integrative single-cell and bulk RNA-seq analysis identifies lactylation-related signature in osteosarcoma. Funct Integr Genomics 2025; 25:60. [PMID: 40072643 DOI: 10.1007/s10142-025-01559-4] [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: 11/28/2024] [Revised: 02/08/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025]
Abstract
Osteosarcoma is the most common bone tumor and a highly aggressive malignant neoplasm. This study aims to elucidate the role of lactylation-related genes (LRGs) in osteosarcoma, with the goal of improving prognostic accuracy and enhancing the efficacy of immunotherapy. Using public datasets, we integrated differential and correlated genes based on single-cell sequencing AUCell scores and performed enrichment analysis and risk model construction on these genes. A total of 277 genes were found to be intricately linked with lactate metabolism. Using the uni-Cox and LASSO algorithm, nine key genes were identified, demonstrating strong predictive power for the prognosis of Osteosarcoma patients. Notably, changes were observed at the levels of immune checkpoints, the tumor microenvironment (TME), drug sensitivity, and immune cell infiltration. This study paves the way for targeted drug interventions, thereby opening avenues for improving clinical outcomes in osteosarcoma.
Collapse
Affiliation(s)
- Zhou Xie
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiao Qu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jun Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yanran Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhao Runhan
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dagang Tang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ningdao Li
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhule Wang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, 400060, China.
| |
Collapse
|
36
|
Guo Y, Mao T, Fang Y, Wang H, Yu J, Zhu Y, Shen S, Zhou M, Li H, Hu Q. Comprehensive insights into potential roles of purinergic P2 receptors on diseases: Signaling pathways involved and potential therapeutics. J Adv Res 2025; 69:427-448. [PMID: 38565403 PMCID: PMC11954808 DOI: 10.1016/j.jare.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/03/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Purinergic P2 receptors, which can be divided into ionotropic P2X receptors and metabotropic P2Y receptors, mediate cellular signal transduction of purine or pyrimidine nucleoside triphosphates and diphosphate. Based on the wide expression of purinergic P2 receptors in tissues and organs, their significance in homeostatic maintenance, metabolism, nociceptive transmission, and other physiological processes is becoming increasingly evident, suggesting that targeting purinergic P2 receptors to regulate biological functions and signal transmission holds significant promise for disease treatment. AIM OF REVIEW This review highlights the detailed mechanisms by which purinergic P2 receptors engage in physiological and pathological progress, as well as providing prospective strategies for discovering clinical drug candidates. KEY SCIENTIFIC CONCEPTS OF REVIEW The purinergic P2 receptors regulate complex signaling and molecular mechanisms in nervous system, digestive system, immune system and as a result, controlling physical health states and disease progression. There has been a significant rise in research and development focused on purinergic P2 receptors, contributing to an increased number of drug candidates in clinical trials. A few influential pioneers have laid the foundation for advancements in the evaluation, development, and of novel purinergic P2 receptors modulators, including agonists, antagonists, pharmaceutical compositions and combination strategies, despite the different scaffolds of these drug candidates. These advancements hold great potential for improving therapeutic outcomes by specifically targeting purinergic P2 receptors.
Collapse
Affiliation(s)
- Yanshuo Guo
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Tianqi Mao
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China
| | - Yafei Fang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hui Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China
| | - Jiayue Yu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yifan Zhu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China
| | - Shige Shen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Mengze Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Huanqiu Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China.
| | - Qinghua Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
37
|
Kostas JC, Brainard CS, Cristea IM. A Primer on Proteomic Characterization of Intercellular Communication in a Virus Microenvironment. Mol Cell Proteomics 2025; 24:100913. [PMID: 39862905 PMCID: PMC11889360 DOI: 10.1016/j.mcpro.2025.100913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Intercellular communication is fundamental to multicellular life and a core determinant of outcomes during viral infection, where the common goals of virus and host for persistence and replication are generally at odds. Hosts rely on encoded innate and adaptive immune responses to detect and clear viral pathogens, while viruses can exploit or disrupt these pathways and other intercellular communication processes to enhance their spread and promote pathogenesis. While virus-induced signaling can result in systemic changes to the host, striking alterations are observed within the cellular microenvironment directly surrounding a site of infection, termed the virus microenvironment (VME). Mechanisms employed by viruses to condition their VMEs are emerging and are critical for understanding the biology and pathologies of viral infections. Recent advances in experimental approaches, including proteomic methods, have enabled study of the VME in unprecedented detail. In this review article, we provide a primer on proteomic approaches used to study how viral infections alter intercellular communication, highlighting the ways in which these approaches have been implemented and the exciting biology they have uncovered. First, we consider the different molecules secreted by an infected cell, including proteins, either soluble or contained within extracellular vesicles, and metabolites. We further discuss the modalities of interactions facilitated by alteration at the cell surface of infected cells, including immunopeptide presentation and interactions with the extracellular matrix. Finally, we review spatial profiling approaches that have allowed distinguishing how specific subpopulations of cells within a VME respond to infection and alter their protein composition, discussing valuable insights these methods have offered.
Collapse
Affiliation(s)
- James C Kostas
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Colter S Brainard
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA.
| |
Collapse
|
38
|
Maino A, Bourova-Flin E, Decaens T, Khochbin S, Macek Jilkova Z, Rousseaux S, Plumas J, Saas P, Chaperot L, Manches O. Identification of immunogenic HLA-A*02:01 epitopes associated with HCC for immunotherapy development. Hepatol Commun 2025; 9:e0659. [PMID: 40008881 PMCID: PMC11868434 DOI: 10.1097/hc9.0000000000000659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND HCC is the most common form of primary liver cancer, and despite recent advances in cancer treatment, it remains associated with poor prognosis and a lack of response to conventional therapies. Immunotherapies have emerged as a promising approach for cancer treatment, especially through the identification of tumor-specific immunogenic epitopes that can trigger a targeted immune response. This study aimed to identify immunogenic epitopes associated with HCC for the development of specific immunotherapies. METHODS We used high-throughput data screening and bioinformatics tools for antigens and epitope selection. The immunogenicity of the selected epitopes was studied after coculture of peripheral blood mononuclear cells obtained from healthy donors or HCC patients with a plasmacytoid dendritic cell line loaded with the selected peptides. Specific CD8+ T cell amplification and functionality were determined by labeling with tetramers and by IFN-γ and CD107a expression (flow cytometry and ELISpot). RESULTS We analyzed the transcriptional gene expression landscape of HCC to screen for a set of 16 ectopically expressed genes in a majority of HCC samples. Epitopes predicted to bind to HLA-A*02:01 with high affinity were further validated for their immunogenicity using the previously described plasmacytoid dendritic cell line in ex vivo CD8+ activation assays using patient immune cells. Three out of the 30 tested epitopes, namely FLWGPRALV (MAGE-A3), FMNKFIYEI (AFP), and KMFHTLDEL (LRRC46), elicited a strong T-cell response, in activation assays, degranulation assays, and IFN-γ secretion assays. CONCLUSIONS These results highlight the potential of these peptides to be considered as targets for immunotherapies. The discovery of such immunogenic epitopes should improve immune-based treatments for liver cancer in combination with the current treatment approach.
Collapse
Affiliation(s)
- Anthony Maino
- EFS, R&D Department, Grenoble, France
- Univ. Grenoble Alpes, INSERM U, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Ekaterina Bourova-Flin
- Univ. Grenoble Alpes, INSERM U, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Thomas Decaens
- Univ. Grenoble Alpes, INSERM U, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
- Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France
| | - Saadi Khochbin
- Univ. Grenoble Alpes, INSERM U, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Zuzana Macek Jilkova
- Univ. Grenoble Alpes, INSERM U, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
- Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France
| | - Sophie Rousseaux
- Univ. Grenoble Alpes, INSERM U, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Joel Plumas
- PDC*line Pharma SAS, R&D Department, Grenoble, France
| | - Philippe Saas
- EFS, R&D Department, Grenoble, France
- Univ. Grenoble Alpes, INSERM U, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Laurence Chaperot
- EFS, R&D Department, Grenoble, France
- Univ. Grenoble Alpes, INSERM U, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Olivier Manches
- EFS, R&D Department, Grenoble, France
- Univ. Grenoble Alpes, INSERM U, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| |
Collapse
|
39
|
Sun P, Liu F, Huo K, Wang J, Cheng Y, Shang S, Ma W, Yu J, Han J. Adiponectin facilitates the cell cycle, inhibits cell apoptosis and induces temozolomide resistance in glioblastoma via the Akt/mTOR pathway. Oncol Lett 2025; 29:127. [PMID: 39807099 PMCID: PMC11726000 DOI: 10.3892/ol.2025.14875] [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: 07/02/2024] [Accepted: 10/22/2024] [Indexed: 01/16/2025] Open
Abstract
Adiponectin (ADN) regulates DNA synthesis, cell apoptosis and cell cycle to participate in the pathology and progression of glioblastoma. The present study aimed to further explore the effect of ADN on temozolomide (TMZ) resistance in glioblastoma and the underlying mechanism of action. Glioblastoma cell lines (U251 and U87-MG cells) were treated with ADN and TMZ at different concentrations; subsequently, 3.0 µg/ml ADN and 1.0 mM TMZ were selected as the optimal concentrations for the experimental conditions. LY294002 (a PI3K inhibitor) was added to ADN or ADN + TMZ-treated glioblastoma cell lines. Cell growth rate was determined using the Cell Counting Kit-8 assay, the apoptotic rate and cell cycle were evaluated using Annexin V/propidium iodide and cell cycle assays, and p-Akt (Thr308), p-Akt (Ser473), Akt, p-mTOR, c-caspase 3, caspase 3, Bax, cyclin B1 and cyclin D1 expression was determined by western blotting. Adiponectin receptor (ADIPOR) 1 and ADIPOR2 were expressed in glioblastoma cell lines. The glioblastoma cell line growth rate was increased by ADN in a concentration- and time-dependent manner. ADN inhibited glioblastoma cell line apoptosis and facilitated cell cycle. Of note, ADN activated the Akt/mTOR pathway and the addition of LY294002 reversed the effect of ADN, indicating that ADN activated the Akt/mTOR pathway to suppress apoptosis and promote cell cycle in glioblastoma cell lines. Notably, TMZ inhibited glioblastoma cell line growth, promoted apoptosis and increased G2 phase cell cycle arrest. However, the addition of ADN reversed the effect of TMZ in glioblastoma cell lines, disclosing that ADN induced TMZ resistance. Markedly, ADN-mediated TMZ resistance was further attenuated by LY294002, suggesting that ADN activated the Akt/mTOR pathway to induce TMZ resistance in glioblastoma cell lines. In conclusion, ADN activated the Akt/mTOR pathway to facilitate cell cycle, inhibit cell apoptosis and induce TMZ resistance in glioblastoma.
Collapse
Affiliation(s)
- Peng Sun
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Fude Liu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jianyi Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yawen Cheng
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenlong Ma
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jia Yu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jianfeng Han
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
40
|
Zhao Y, Yang J, Xu S, Wang Y, Bo J. The pathogenesis of B-cell non-hodgkin lymphoma associated with HBV (hepatitis B virus) infection is regulated by c-Myc/PD-L1 signaling pathway. Glob Med Genet 2025; 12:100001. [PMID: 39925444 PMCID: PMC11800311 DOI: 10.1016/j.gmg.2024.100001] [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] [Indexed: 02/11/2025] Open
Abstract
Background HBV is closely associated with the incidence of B-NHL (B-cell non-hodgkin lymphoma). This project intends to establish HBV infection-induced B-NHL cells and animal models to clarify the mutual mechanism of HBV infection and B-NHL pathogenesis. Methods The relationship between HBV and B-NHL was studied based on the HBV infection model, which included CTC cells and HBV transgenic mice. Moreover, differential expression analysis of transcriptome profiling was performed to confirm the mechanism. Results The HBsAg expression and HBV-DNA could be found in tumor tissues of HBV group, but negative in the control group. Moreover, there were clearly differences between the two groups in the transcriptome of tumor tissues and CTC. HBsAg significantly promoted lymphocytes associated with c-Myc and PD-L1. Conclusion The promoted effect induced by HBsAg in lymphocytes was associated with PD-L1 mediated by c-Myc.
Collapse
Affiliation(s)
- Yu Zhao
- Senior Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, No 8 East Street, Fengtai District, Beijing 100071, China
| | - Jian Yang
- Clinic of Yongding Road, Southern Medical Branch of PLA General Hospital, No 27 Taiping Road, Haidian District, Beijing 100036, China
| | - Sai Xu
- Senior Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, No 8 East Street, Fengtai District, Beijing 100071, China
| | - Ying Wang
- Clinic of Yongding Road, Southern Medical Branch of PLA General Hospital, No 27 Taiping Road, Haidian District, Beijing 100036, China
| | - Jian Bo
- Senior Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, No 8 East Street, Fengtai District, Beijing 100071, China
| |
Collapse
|
41
|
Daya T, Breytenbach A, Gu L, Kaur M. Cholesterol metabolism in pancreatic cancer and associated therapeutic strategies. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159578. [PMID: 39542394 DOI: 10.1016/j.bbalip.2024.159578] [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: 07/24/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Pancreatic cancer remains one of the most lethal cancers due to late diagnosis and high chemoresistance. Despite recent progression in the development of chemotherapies, immunotherapies, and potential nanoparticles-based approaches, the success rate of therapeutic response is limited which is further compounded by cancer drug resistance. Understanding of emerging biological and molecular pathways causative of pancreatic cancer's aggressive and chemoresistance is vital to improve the effectiveness of existing therapeutics and to develop new therapies. One such under-investigated and relatively less explored area of research is documenting the effect that lipids, specifically cholesterol, and its metabolism, impose on pancreatic cancer. Dysregulated cholesterol metabolism has a profound role in supporting cellular proliferation, survival, and promoting chemoresistance and this has been well established in various other cancers. Thus, we aimed to provide an in-depth review focusing on the significance of cholesterol metabolism in pancreatic cancer and relevant genes at play, molecular processes contributing to cellular cholesterol homeostasis, and current research efforts to develop new cholesterol-targeting therapeutics. We highlight the caveats, weigh in different experimental therapeutic strategies, and provide possible suggestions for future research highlighting cholesterol's importance as a therapeutic target against pancreatic cancer resistance and cancer progression.
Collapse
Affiliation(s)
- Tasvi Daya
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Andrea Breytenbach
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Liang Gu
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa.
| |
Collapse
|
42
|
Zhang C, Chen X, Yan C, Lv R, An S, Gao Y, Huang T, Deng W. HBX Multi-Mutations Combined With Traditional Screening Indicators to Establish a Nomogram Contributes to Precisely Stratify the High-Risk Population of Hepatocellular Carcinoma. Cancer Med 2025; 14:e70748. [PMID: 40042093 PMCID: PMC11880911 DOI: 10.1002/cam4.70748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 02/10/2025] [Accepted: 02/26/2025] [Indexed: 05/12/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most prevalent malignant tumors, often diagnosed at an advanced stage with limited treatment options and a poor prognosis. The present study aimed to identify the risk factors (RFs) for HCC and develop a nomogram incorporating dominant HBX mutations to predict the risk of HCC occurrence in high-risk (HR) populations. METHODS We collected early HCC screening and monitoring factors from cohorts of HCC patients and HR populations, including gender, age, AFP, ALT, as well as hepatitis B virus (HBV) infection and mutation indicators such as hepatitis B surface antigen (HBsAg), HBV DNA replication level, HBV genotype, and high-frequency mutations in HBX. Independent predictive factors for HCC onset were determined through both univariate and multivariate logistic regression analyses. Two nomograms with and without HBX mutation data were established to predict the risk of HCC incidence in HR populations, and their performance was evaluated using calibration curves, receiver operating characteristic (ROC) curves, as well as decision curve analysis (DCA). RESULTS A total of 312 participants were included. Independent RFs for HCC onset were identified as A1762T+G1764A multi-mutations, T1753C/G/A+A1762T+G1764A multi-mutations, and ALT > 40 U/L. The area under the curve (AUC) of the diagnostic nomogram with HBX mutation data was 0.835 in the training set and 0.869 in the testing set for the nomogram. Besides, the AUC of the diagnostic nomogram without HBX mutation data in the training set was 0.798 and 0.818 in the testing set. The calibration curve together with DCA indicated that the nomogram containing HBX mutation data had better predictive performance. CONCLUSIONS The established nomograms predicted the risk of HCC occurrence in HR populations with good accuracy, providing a valuable reference for precise stratification of HR populations and HCC screening.
Collapse
Affiliation(s)
- Chao‐Jun Zhang
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningGuangxiChina
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningGuangxiPeople's Republic of China
| | - Xiao‐Mei Chen
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Chang Yan
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningGuangxiPeople's Republic of China
| | - Rui‐Bo Lv
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Sanchun An
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Yun‐Xin Gao
- Guangdong Forevergen Medical Technology Co LtdFoshanGuangdongChina
| | - Tian‐Ren Huang
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningGuangxiChina
- Guangxi Cancer Molecular Medicine Engineering Research CenterNanningGuangxiChina
| | - Wei Deng
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningGuangxiChina
- Guangxi Cancer Molecular Medicine Engineering Research CenterNanningGuangxiChina
| |
Collapse
|
43
|
Bloom M, Podder S, Dang H, Lin D. Advances in Immunotherapy in Hepatocellular Carcinoma. Int J Mol Sci 2025; 26:1936. [PMID: 40076561 PMCID: PMC11900920 DOI: 10.3390/ijms26051936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Over the past several years, the therapeutic landscape for patients with advanced, unresectable, or metastatic hepatocellular carcinoma has been transformed by the incorporation of checkpoint inhibitor immunotherapy into the treatment paradigm. Frontline systemic treatment options have expanded beyond anti-angiogenic tyrosine kinase inhibitors, such as sorafenib, to a combination of immunotherapy approaches, including atezolizumab plus bevacizumab and durvalumab plus tremelimumab, both of which have demonstrated superior response and survival to sorafenib. Additionally, combination treatments with checkpoint inhibitors and tyrosine kinase inhibitors have been investigated with variable success. In this review, we discuss these advances in systemic treatment with immunotherapy, with a focus on understanding both the underlying biology and mechanism of these strategies and their efficacy outcomes in clinical trials. We also review challenges in identifying predictive biomarkers of treatments and discuss future directions with novel immunotherapy targets.
Collapse
Affiliation(s)
- Matthew Bloom
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA;
| | - Sourav Podder
- Department of Surgery, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA; (S.P.); (H.D.)
| | - Hien Dang
- Department of Surgery, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA; (S.P.); (H.D.)
| | - Daniel Lin
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA;
| |
Collapse
|
44
|
Shi M, Ning Z. In vivo and in vitro investigations of schisandrin B against angiotensin II induced ferroptosis and atrial fibrosis by regulation of the SIRT1 pathway. Sci Rep 2025; 15:6200. [PMID: 39979353 PMCID: PMC11842858 DOI: 10.1038/s41598-025-89895-0] [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: 11/21/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
Schisandrin B (Sch B) derived from Schisandra chinensis, is known for its anti-inflammatory and anti-microbial properties. The study aimed to explore Sch B's protective roles and underlying mechanisms in angiotensin II (Ang II) - induced ferroptosis, atrial fibrosis, and AF using both in vivo and in vitro models. AF mice model generated induced by Ang II and established an in vitro model using the HL-1 cell line induced by Ang II. We assessed atrial fibrosis through histological analysis and oxidative stress analysis. We employed RT-qPCR and Western blot techniques to evaluate mRNA and protein expression. Sch B significantly attenuated Ang II-induced AF development, atrial apoptosis, and myocardial injury-related molecules, including CK-MB and LDH. Relative DHE intensity, MDA, NOX2, and NOX4 increased significantly, and SOD and CAT levels decreased markedly in Ang II-induced mice. Sch B treatment could inhibit atrial ROS production and oxidative stress in Ang II-infused mice. In addition, Sch B showed cardioprotective effects in Ang II-infused HL-1 cells. Sch B significantly reduced pro-inflammatory cytokines, including IL-1β, TNF-α, and IL-6, restored by EX527 (SIRT1 inhibitor). Sch B inhibited intracellular ROS generation and oxidative stress in HL-1 cells, which were restored by Ex-527. Furthermore, Sch B decreased the increase in Fe2 + concentration caused by Ang II infusion, which was recovered by Ex-527. Sch B markedly increased the expression of SIRT1, SLC7A11, GPX4 and FTH1 while reducing the expression patterns by Ex-527 treatment. Our experimental data suggest that Sch B protects against Ang II-induced ferroptosis, atrial fibrosis, and AF by activating SIRT1 in vivo and in vitro.
Collapse
Affiliation(s)
- Mengqing Shi
- Graduate School, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai, 201203, China
| | - Zhongping Ning
- Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai, 201203, China.
- Department of Cardiology, Shanghai Pudong New Area Zhoupu Hospital (Shanghai Health Medical College Affiliated Zhoupu Hospital), No.1500 Zhou Yuan Road, Pudong New District, Shanghai, 201318, China.
| |
Collapse
|
45
|
Song Y, Wang Y, Man J, Xu Y, Zhou G, Shen W, Chao Y, Yang K, Pei P, Hu L. Chimeric Antigen Receptor Cells Solid Tumor Immunotherapy Assisted by Biomaterials Tools. ACS APPLIED MATERIALS & INTERFACES 2025; 17:10246-10264. [PMID: 39903799 DOI: 10.1021/acsami.4c20275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Chimeric antigen receptor (CAR) immune cell therapies have revolutionized oncology, particularly in hematological malignancies, yet their efficacy against solid tumors remains limited due to challenges such as dense stromal barriers and immunosuppressive microenvironments. With advancements in nanobiotechnology, researchers have developed various strategies and methods to enhance the CAR cell efficacy in solid tumor treatment. In this Review, we first outline the structure and mechanism of CAR-T (T, T cell), CAR-NK (NK, natural killer), and CAR-M (M, macrophage) cell therapies and deeply analyze the potential of these cells in the treatment of solid tumors and the challenges they face. Next, we explore how biomaterials can optimize these treatments by improving the tumor microenvironment, controlling CAR cell release, promoting cell infiltration, and enhancing efficacy. Finally, we summarize the current challenges and potential solutions, emphasize the effective combination of biomaterials and CAR cell therapy, and look forward to its future clinical application and treatment strategies. This Review provides important theoretical perspectives and practical guidance for the future development of more effective solid tumor treatment strategies.
Collapse
Affiliation(s)
- Yujie Song
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yifan Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jianping Man
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yihua Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Wenhao Shen
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Yu Chao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pei Pei
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
46
|
Chen JG, Kensler TW, Zhu J, Zhu YR, Wang JB, Lu JH, Muñoz A, Groopman JD. Profound primary prevention of liver cancer following a natural experiment in China: A 50-year perspective and public health implications. Int J Cancer 2025; 156:756-763. [PMID: 39305480 DOI: 10.1002/ijc.35198] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 12/21/2024]
Abstract
Liver cancer causes upwards of 1 million cancer deaths annually and is projected to rise by at least 55% over the next 15 years. Two of the major risk factors contributing to liver cancer have been well documented by multiple epidemiologic studies and the hepatitis B virus (HBV) and aflatoxin show a synergy that increases by more than 8-fold the risk of liver cancer relative to HBV alone. Using the population-based cancer registry established by the Qidong Liver Cancer Institute in 1972 and aflatoxin-specific biomarkers, we document that reduction of aflatoxin exposure has likely contributed to a nearly 70% decline in age-standardized liver cancer incidence over the past 30 years despite an unchanging prevalence of HBV infection in cases. A natural experiment of economic reform in the 1980s drove a rapid switch from consumption of heavily contaminated corn to minimally, if any, contaminated rice and subsequent dietary diversity. Aflatoxin consumption appears to accelerate the time to liver cancer diagnosis; lowering exposure to this carcinogen adds years of life before a cancer diagnosis. Thus, in 1990 the median age of diagnosis was 48 years, while increasing to 67 years by 2021. These findings have important translational public health implications since up to 5 billion people worldwide might be routinely exposed to dietary aflatoxin, especially in societies using corn as the staple food. Interventions against aflatoxin are an achievable outcome leading to a reduction in liver cancer incidence and years of delay of its nearly always fatal diagnosis.
Collapse
Affiliation(s)
- Jian-Guo Chen
- Qidong Liver Cancer Institute, Qidong People's Hospital, Affiliated Qidong Hospital of Nantong University, Nantong, China
| | - Thomas W Kensler
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jian Zhu
- Qidong Liver Cancer Institute, Qidong People's Hospital, Affiliated Qidong Hospital of Nantong University, Nantong, China
| | - Yuan-Rong Zhu
- Qidong Liver Cancer Institute, Qidong People's Hospital, Affiliated Qidong Hospital of Nantong University, Nantong, China
| | - Jin-Bing Wang
- Qidong Liver Cancer Institute, Qidong People's Hospital, Affiliated Qidong Hospital of Nantong University, Nantong, China
| | - Jian-Hua Lu
- Qidong Liver Cancer Institute, Qidong People's Hospital, Affiliated Qidong Hospital of Nantong University, Nantong, China
| | - Alvaro Muñoz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - John D Groopman
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
47
|
Hu C, Chen Y, Yin X, Xu R, Yin C, Wang C, Zhao Y. Pancreatic endocrine and exocrine signaling and crosstalk in physiological and pathological status. Signal Transduct Target Ther 2025; 10:39. [PMID: 39948335 PMCID: PMC11825823 DOI: 10.1038/s41392-024-02098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/20/2024] [Accepted: 12/03/2024] [Indexed: 02/16/2025] Open
Abstract
The pancreas, an organ with dual functions, regulates blood glucose levels through the endocrine system by secreting hormones such as insulin and glucagon. It also aids digestion through the exocrine system by secreting digestive enzymes. Complex interactions and signaling mechanisms between the endocrine and exocrine functions of the pancreas play a crucial role in maintaining metabolic homeostasis and overall health. Compelling evidence indicates direct and indirect crosstalk between the endocrine and exocrine parts, influencing the development of diseases affecting both. From a developmental perspective, the exocrine and endocrine parts share the same origin-the "tip-trunk" domain. In certain circumstances, pancreatic exocrine cells may transdifferentiate into endocrine-like cells, such as insulin-secreting cells. Additionally, several pancreatic diseases, including pancreatic cancer, pancreatitis, and diabetes, exhibit potential relevance to both endocrine and exocrine functions. Endocrine cells may communicate with exocrine cells directly through cytokines or indirectly by regulating the immune microenvironment. This crosstalk affects the onset and progression of these diseases. This review summarizes the history and milestones of findings related to the exocrine and endocrine pancreas, their embryonic development, phenotypic transformations, signaling roles in health and disease, the endocrine-exocrine crosstalk from the perspective of diseases, and potential therapeutic targets. Elucidating the regulatory mechanisms of pancreatic endocrine and exocrine signaling and provide novel insights for the understanding and treatment of diseases.
Collapse
Grants
- National High Level Hospital Clinical Research Funding (2022, 2022-PUMCH-D-001, to YZ), CAMS Innovation Fund for Medical Sciences (2021, 2021-I2M-1-002, to YZ), National Nature Science Foundation of China (2021, 82102810, to CW, the Fundamental Research Funds for the Central Universities(3332023123)
- cNational High Level Hospital Clinical Research Funding (2022, 2022-PUMCH-D-001, to YZ), CAMS Innovation Fund for Medical Sciences (2021, 2021-I2M-1-002, to YZ), National Nature Science Foundation of China (2021, 82102810, to CW, the Fundamental Research Funds for the Central Universities(3332023123)
Collapse
Affiliation(s)
- Chenglin Hu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Chenxue Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Chengcheng Wang
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China.
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, PR China.
- Institute of Clinical Medicine, Peking Union Medical College Hospital, Beijing, PR China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China.
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, PR China.
| |
Collapse
|
48
|
Chen JG, Zhu YR, Qian GS, Wang JB, Lu JH, Kensler TW, Jacobson LP, Muñoz A, Groopman JD. Fifty Years of Aflatoxin Research in Qidong, China: A Celebration of Team Science to Improve Public Health. Toxins (Basel) 2025; 17:79. [PMID: 39998096 PMCID: PMC11860843 DOI: 10.3390/toxins17020079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
The Qidong Liver Cancer Institute (QDLCI) and the Qidong Cancer Registry were established in 1972 with input from doctors, other medical practitioners, and non-medical investigators arriving from urban centers such as Shanghai and Nanjing. Medical teams were established to quantify the extent of primary liver cancer in Qidong, a corn-growing peninsula on the north side of the Yangtze River. High rates of liver cancer were documented and linked to several etiologic agents, including aflatoxins. Local corn, the primary dietary staple, was found to be consistently contaminated with high levels of aflatoxins, and bioassays using this corn established its carcinogenicity in ducks and rats. Observational studies noted a positive association between levels of aflatoxin in corn and incidence of liver cancer across townships. Biomarker studies measuring aflatoxin B1 and its metabolite aflatoxin M1 in biofluids reflected the exposures. Approaches to decontamination of corn from aflatoxins were also studied. In 1993, investigators from Johns Hopkins University were invited to visit the QDLCI to discuss chemoprevention studies in some townships. A series of placebo-controlled clinical trials were conducted using oltipraz (a repurposed drug), chlorophyllin (an over-the-counter drug), and beverages prepared from 3-day-old broccoli sprouts (rich in the precursor phytochemical for sulforaphane). Modulation of biomarkers of aflatoxin DNA and albumin adducts established proof of principle for the efficacy of these agents in enhancing aflatoxin detoxication. Serendipitously, by 2012, aflatoxin exposures quantified using biomarker measurements documented a many hundred-fold reduction. In turn, the Cancer Registry documents that the age-standardized incidence rate of liver cancer is now 75% lower than that seen in the 1970s. This reduction is seen in Qidongese who have never received the hepatitis B vaccination. Aflatoxin mitigation driven by economic changes switched the dietary staple of contaminated corn to rice coupled with subsequent dietary diversity leading to lower aflatoxin exposures. This 50-year effort to understand the etiology of liver cancer in Qidong provides the strongest evidence for aflatoxin mitigation as a public health strategy for reducing liver cancer burden in exposed, high-risk populations. Also highlighted are the challenges and successes of international team science to solve pressing public health issues.
Collapse
Affiliation(s)
- Jian-Guo Chen
- Department of Epidemiology, Qidong Liver Cancer Institute, Qidong People’s Hospital, Affiliated Qidong Hospital of Nantong University, Qidong 226200, China; (Y.-R.Z.); (J.-B.W.); (J.-H.L.)
| | - Yuan-Rong Zhu
- Department of Epidemiology, Qidong Liver Cancer Institute, Qidong People’s Hospital, Affiliated Qidong Hospital of Nantong University, Qidong 226200, China; (Y.-R.Z.); (J.-B.W.); (J.-H.L.)
| | - Geng-Sun Qian
- Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai 200032, China;
| | - Jin-Bing Wang
- Department of Epidemiology, Qidong Liver Cancer Institute, Qidong People’s Hospital, Affiliated Qidong Hospital of Nantong University, Qidong 226200, China; (Y.-R.Z.); (J.-B.W.); (J.-H.L.)
| | - Jian-Hua Lu
- Department of Epidemiology, Qidong Liver Cancer Institute, Qidong People’s Hospital, Affiliated Qidong Hospital of Nantong University, Qidong 226200, China; (Y.-R.Z.); (J.-B.W.); (J.-H.L.)
| | - Thomas W. Kensler
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | - Lisa P. Jacobson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (L.P.J.); (A.M.)
| | - Alvaro Muñoz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (L.P.J.); (A.M.)
| | - John D. Groopman
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| |
Collapse
|
49
|
Ma X, Huang T, Chen X, Li Q, Liao M, Fu L, Huang J, Yuan K, Wang Z, Zeng Y. Molecular mechanisms in liver repair and regeneration: from physiology to therapeutics. Signal Transduct Target Ther 2025; 10:63. [PMID: 39920130 PMCID: PMC11806117 DOI: 10.1038/s41392-024-02104-8] [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: 11/08/2023] [Revised: 09/02/2024] [Accepted: 12/12/2024] [Indexed: 02/09/2025] Open
Abstract
Liver repair and regeneration are crucial physiological responses to hepatic injury and are orchestrated through intricate cellular and molecular networks. This review systematically delineates advancements in the field, emphasizing the essential roles played by diverse liver cell types. Their coordinated actions, supported by complex crosstalk within the liver microenvironment, are pivotal to enhancing regenerative outcomes. Recent molecular investigations have elucidated key signaling pathways involved in liver injury and regeneration. Viewed through the lens of metabolic reprogramming, these pathways highlight how shifts in glucose, lipid, and amino acid metabolism support the cellular functions essential for liver repair and regeneration. An analysis of regenerative variability across pathological states reveals how disease conditions influence these dynamics, guiding the development of novel therapeutic strategies and advanced techniques to enhance liver repair and regeneration. Bridging laboratory findings with practical applications, recent clinical trials highlight the potential of optimizing liver regeneration strategies. These trials offer valuable insights into the effectiveness of novel therapies and underscore significant progress in translational research. In conclusion, this review intricately links molecular insights to therapeutic frontiers, systematically charting the trajectory from fundamental physiological mechanisms to innovative clinical applications in liver repair and regeneration.
Collapse
Affiliation(s)
- Xiao Ma
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiangzheng Chen
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qian Li
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mingheng Liao
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Fu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiwei Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yong Zeng
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
50
|
Wei X, Xiong X, Chen Z, Chen B, Zhang C, Zhang W. MicroRNA155 in non-small cell lung cancer: a potential therapeutic target. Front Oncol 2025; 15:1517995. [PMID: 39963112 PMCID: PMC11830606 DOI: 10.3389/fonc.2025.1517995] [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: 10/27/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Lung cancer (LC) is the second most commonly diagnosed cancer among both men and women, and it stands as the leading cause of cancer-related mortality, characterized by high rates of morbidity and mortality. Among its subtypes, non-small cell lung cancer (NSCLC) is the most prevalent and one of the most challenging malignant tumors to treat. To date, various therapeutic approaches, including surgery, radiotherapy, and chemotherapy, have been employed in the management of lung cancer; however, due to its aggressive nature, the survival rates remain low. Consequently, exploring novel treatment strategies is of paramount importance. MicroRNAs (miRNAs), a large family of non-coding RNAs, play crucial roles in regulating several key biological processes, including cell proliferation, differentiation, inflammation, and apoptosis. Among these, microRNA155(miR-155) is one of the most conserved and versatile miRNAs, predominantly overexpressed in various diseases, including malignant tumors. This review elucidates the biological functions and roles of miR-155 in NSCLC and discusses its potential significance as a therapeutic target for future research directions and clinical applications.
Collapse
Affiliation(s)
- Xiangju Wei
- The First Clinical College, Xuzhou Medical University, Xuzhou, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xianmin Xiong
- The First Clinical College, Xuzhou Medical University, Xuzhou, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ze Chen
- The First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Bi Chen
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Cantang Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wenhui Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|