201
|
Chen Y, Li K, Du H, Yao Y, Xie D, Zhou Z. Breaking Barriers in Oncology: Harnessing Sonodynamic Therapy for Enhanced Tumor Metabolism Regulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2502323. [PMID: 40317653 DOI: 10.1002/smll.202502323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/01/2025] [Indexed: 05/07/2025]
Abstract
The recent booming development of sonometabolism regulation in controlling the tumor microenvironment (TME) has opened a new research area to identify innovative approaches against cancer. The aim of this review is to highlight the potentials and advantages of sonodynamic therapy (SDT) in antitumor nanotherapies, specifically, delineating the progress made in SDT concerning the regulation of TME metabolism which encompasses factors such as hypoxia, redox balance, autophagy, immunosuppression, ion homeostasis, and other metabolic processes. By focusing on both tumor cell metabolism and TME dynamics, a wide range of SDT strategies that have demonstrated great therapeutic effectiveness by targeting the metabolic functions inherent to TME are summarized. In conclusion, this review offers valuable insights for researchers involved in SDT-based antitumor therapeutic strategies, with the aim of advancing the development of antitumor SDT methodologies in future research.
Collapse
Affiliation(s)
- Yangmengfan Chen
- Department of Orthopedics and Research Institute of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kun Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Hao Du
- Department of Orthopedics and Research Institute of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yangcheng Yao
- Center for Reproductive Medicine, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Zongke Zhou
- Department of Orthopedics and Research Institute of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
202
|
Deng J, Du Z, Li L, Zhu M, Zhao H. Phase separation in DNA repair: orchestrating the cellular response to genomic stability. PeerJ 2025; 13:e19402. [PMID: 40330699 PMCID: PMC12051939 DOI: 10.7717/peerj.19402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
DNA repair is a hierarchically organized, spatially and temporally regulated process involving numerous repair factors that respond to various types of damage. Despite decades of research, the mechanisms by which these factors are recruited to and depart from repair sites have been a subject of intrigue. Recent advancements in the field have increasingly highlighted the role of phase separation as a critical facilitator of the efficiency of DNA repair. This review emphasizes how phase separation enhances the concentration and coordination of repair factors at damage sites, optimizing repair efficiency. Understanding how dysregulation of phase separation can impair DNA repair and alter nuclear organization, potentially leading to diseases such as cancer and neurodegenerative disorders, is crucial. This manuscript provides a comprehensive understanding of the pivotal role of phase separation in DNA repair, sheds light on the current research, and suggests potential future directions for research and therapeutic interventions.
Collapse
Affiliation(s)
- Juxin Deng
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Zhaoyang Du
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lei Li
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical University, Institute of Emergency and Critical Care Medicine, Bengbu, Anhui, China
| | - Min Zhu
- School of Life Science, Anhui Agriculture University, Hefei, Anhui, China
| | - Hongchang Zhao
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical University, Institute of Emergency and Critical Care Medicine, Bengbu, Anhui, China
| |
Collapse
|
203
|
Xu Y, Hu R, Zhang X. Recent Advances in Reactive Oxygen Species -Mediated Near-Infrared Organic Long-Persistent Luminescence Imaging. Chem Asian J 2025; 20:e202401918. [PMID: 39945087 DOI: 10.1002/asia.202401918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/08/2025] [Indexed: 03/14/2025]
Abstract
Organic luminophores have found extensive applications in cellular and in vivo fluorescence imaging. However, their efficacy is often hindered by formidable challenges, including a low signal-to-noise ratio (SNR), susceptibility to false-positive signals, limited tissue penetration depth, and autofluorescence arising from non-negligible background interference. The emergence of near-infrared (NIR) afterglow imaging has addressed these problems. Organic afterglow imaging distinguishes by its unique capacity to emit light long after the cessation of external excitation, thereby exhibiting extraordinary persistence in luminescence. The integration of deep tissue penetration with prolonged luminescence in NIR organic long-persistent luminescent materials confers a distinct advantage for in vivo biological imaging, effectively minimizing the confounding effects of autofluorescence while enhancing spatial resolution for imaging in deep tissues, which is favorable for biosensing. In this review, we present a comprehensive summary of recent advancements in reactive oxygen species (ROS)-mediated NIR organic afterglow imaging, positioning this emerging technique as an exceptionally promising approach for in vivo biosensing, biological imaging, imaging-guided surgery, and therapeutic applications. Furthermore, we critically examine the challenges facing this field and propose future avenues for its continued evolution and refinement.
Collapse
Affiliation(s)
- Yan Xu
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, Yunnan, 650500, P.R. China
| | - Rong Hu
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, Yunnan, 650500, P.R. China
| | - Xiaobing Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theronastics, Hunan University, Changsha, 410082, China
| |
Collapse
|
204
|
Gong Z, Cheng C, Sun C, Cheng X. Harnessing engineered extracellular vesicles for enhanced therapeutic efficacy: advancements in cancer immunotherapy. J Exp Clin Cancer Res 2025; 44:138. [PMID: 40317075 PMCID: PMC12048990 DOI: 10.1186/s13046-025-03403-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025] Open
Abstract
Extracellular vesicles (EVs), particularly engineered variants, have emerged as promising tools in cancer immunotherapy due to their inherent ability to modulate immune responses and deliver therapeutic agents with high specificity and minimal toxicity. These nanometer-sized vesicles, which include exosomes (Exos) and other subtypes, naturally participate in intercellular communication and are capable of carrying a diverse range of bioactive molecules, including proteins, lipids, RNAs, and metabolites. Recent advancements in the biogenesis of engineered EVs, such as strategies to modify their surface characteristics and cargo, have significantly expanded their potential as effective vehicles for targeted cancer therapies. Tailoring the contents of EVs, such as incorporating immunomodulatory molecules or gene-editing tools (GETs), has shown promising outcomes in enhancing anti-tumor immunity and overcoming the immunosuppressive tumor microenvironment (TME). Moreover, optimizing delivery mechanisms, through both passive and active targeting strategies, is crucial for improving the clinical efficacy of EV-based therapies. This review provides an overview of recent developments in the engineering of EVs for cancer immunotherapy, focusing on their biogenesis, methods of content customization, and innovations in cargo delivery. Additionally, the review addresses the challenges associated with the clinical translation of EV-based therapies, such as issues related to scalability, safety, and targeted delivery. By offering insights into the current state of the field and identifying key areas for future research, this review aims to advance the application of engineered EVs in cancer treatment.
Collapse
Affiliation(s)
- Zheng Gong
- Department of Radiology, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang, 110004, China
| | - Cheng Cheng
- Department of Cardiology, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang, 110004, China
| | - Chaonan Sun
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning, 110042, China.
| | - Xiaoli Cheng
- Department of Cardiology, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang, 110004, China.
| |
Collapse
|
205
|
Mai Z, Chen X, Lu Y, Zheng J, Lin Y, Lin P, Zheng Y, Zhou Z, Xu R, Guo B, Cui L, Zhao X. Orchestration of immunoregulatory signaling ligand and receptor dynamics by mRNA modifications: Implications for therapeutic potential. Int J Biol Macromol 2025; 310:142987. [PMID: 40210040 DOI: 10.1016/j.ijbiomac.2025.142987] [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: 01/15/2025] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
RNA modifications are pivotal regulators of gene expression, significantly influencing immune responses by modulating the stability and translation of mRNAs encoding key immunoregulatory ligands and receptors. Among these modifications, N6-methyladenosine (m6A) is the most abundant and well-characterized, orchestrating immune evasion, T-cell exhaustion, and cytokine production by dynamically regulating transcripts such as PD-L1, IFN-γ, and TGF-β. These modifications critically impact the function and availability of proteins essential for maintaining immune homeostasis and shaping adaptive immune responses. This review comprehensively examines established and emerging roles of mRNA modifications in regulating immunoregulatory signaling, including co-inhibitory and co-stimulatory molecules, chemokines, cytokines, and transforming growth factor-β. We highlight how m6A writers, erasers, and readers finely regulate immune checkpoints and inflammatory pathways across cancer, infection, and autoimmune diseases. Furthermore, the review provides a critical analysis of current discrepancies in the field, emphasizing factors contributing to inconsistencies and offering insights into the complex nature of epigenetic regulation. Challenges and limitations in this rapidly evolving area are also discussed. Advancing detection technologies and developing specific inhibitors targeting RNA-modifying proteins will be crucial for precisely modulating immune responses, paving the way for innovations in precision medicine and immunotherapy.
Collapse
Affiliation(s)
- Zizhao Mai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Xu Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Yucheng Zheng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Zihao Zhou
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Rongwei Xu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Bing Guo
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China; School of Dentistry, University of California, Los Angeles, Los Angeles 90095, CA, USA.
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China.
| |
Collapse
|
206
|
Huang L, Ming J, Wang Z, Wu J, Yun B, Liang A, Fan Y, Zhang F. Noninvasively Real-Time Monitoring In-Vivo Immune Cell and Tumor Cell Interaction by NIR-II Nanosensor. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2420329. [PMID: 40150972 DOI: 10.1002/adma.202420329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/18/2025] [Indexed: 03/29/2025]
Abstract
Immunocytotherapy holds significant promise as a novel cancer treatment, but its effectiveness is often hindered by delayed responses, requiring evaluations every 2-3 weeks based on current diagnostic methods. Early assessment of immune cell-tumor cell interactions could provide more timely insights into therapeutic efficacy, enabling adjustments to treatment plans. In this study, a noninvasive nanosensor (C8R-DSNP) for real-time monitoring of in vivo immune cell activities in the second near-infrared long-wavelength (NIR-II-L) window (1500-1900 nm), which offers deep tissue transparency, is reported. The C8R-DSNP responds rapidly to caspase-8, a key apoptotic signaling molecule generated during interactions between natural killer (NK-92) cells and tumor cells. Using ratiometric NIR-II-L fluorescence imaging, dynamic in vivo observations of NK-92 cells' engagement with tumor cells in a mouse model are captured. These results demonstrate tumor cells apoptosis that happens as early as 4.5 h after NK-92 cells infusion. Additionally, in vitro urine imaging confirmed the initiation of apoptosis via cleaved fluorescent small molecules, while single-cell tracking within blood vessels and tumors further elucidated immune cell dynamics. This real-time NIR-II-L monitoring approach offers valuable insights for optimizing immunocytotherapy strategies.
Collapse
Affiliation(s)
- Liwen Huang
- Department of Hematology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, P. R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, 200433, P. R. China
| | - Jiang Ming
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, 200433, P. R. China
| | - Zhihua Wang
- Department of Hematology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, P. R. China
| | - Jiaxin Wu
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, 200433, P. R. China
| | - Baofeng Yun
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, 200433, P. R. China
| | - Aibin Liang
- Department of Hematology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, P. R. China
| | - Yong Fan
- Department of Hematology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, P. R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, 200433, P. R. China
| | - Fan Zhang
- Department of Hematology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, P. R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, 200433, P. R. China
| |
Collapse
|
207
|
Schmitz C, Sforca ML, Maraschin M. Metabolomic Analysis and Computational Biology Reveal the Potential Metabolic Pathways for Improvement of Fucoxanthin and Eicosapentaenoic Acid (EPA) Biosynthesis in Phaeodactylum tricornutum. Appl Biochem Biotechnol 2025; 197:3132-3151. [PMID: 39821501 DOI: 10.1007/s12010-024-05136-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2024] [Indexed: 01/19/2025]
Abstract
This work aims to provide a basis for the enhancement of fucoxanthin (FCX) and eicosapentaenoic acid (EPA) biosynthesis in the microalga Phaeodactylum tricornutum using metabolomics and computational biology. To achieve this, both targeted (UHPLC and GC-FID) and untargeted (FTIR and NMR) analyses were conducted throughout various stages of cell cultivation. Targeted analyses revealed that EPA concentrations peaked at the end of the logarithmic growth phase, while fucoxanthin levels remained consistent from the onset of this phase through to the stationary phase. Untargeted analyses provided metabolic profiles by correlating FTIR absorbance bands with functional groups. When combined with cultivation strategies designed to improve EPA and FCX content, the optimal time for harvesting cells was identified as the end of the logarithmic phase. NMR further highlighted potentially key metabolic pathways for optimizing EPA and FCX production in Phaeodactylum tricornutum, particularly those involved in glyoxylate and dicarboxylate metabolism.
Collapse
Affiliation(s)
- Caroline Schmitz
- Food Biotechnology Laboratory, University of Vale do Taquari - Univates, Lajeado, Rio Grande Do Sul, Brazil.
- Plant Morphogenesis and Biochemistry Laboratory, Federal University of Santa Catarina - UFSC, Florianopolis, Santa Catarina, Brazil.
| | - Maurício Luis Sforca
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, São Paulo, Brazil
| | - Marcelo Maraschin
- Plant Morphogenesis and Biochemistry Laboratory, Federal University of Santa Catarina - UFSC, Florianopolis, Santa Catarina, Brazil
| |
Collapse
|
208
|
Weiskirchen R, Weiskirchen S, Grassi C, Scaggiante B, Grassi M, Tierno D, Biasin A, Truong NH, Minh TD, Cemazar M, Pastorin G, Tonon F, Grassi G. Recent advances in optimizing siRNA delivery to hepatocellular carcinoma cells. Expert Opin Drug Deliv 2025; 22:729-745. [PMID: 40126051 DOI: 10.1080/17425247.2025.2484287] [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: 01/13/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Hepatocellularcarcinoma (HCC), the primary form of liver cancer, is the second leading cause of cancer-related deaths worldwide. Current therapies have limited effectiveness, particularly in advanced stages of the disease, highlighting the need for innovative treatment options. Small-interfering RNA(siRNA) molecules show great promise as a therapeutic solution since they can inhibit the expression of genes promoting HCC growth. Their cost-effective synthesis has further encouraged their potential use as novel drugs. However, siRNAs are vulnerable to degradation in biological environments, necessitating protective delivery systems. Additionally, targeted delivery to HCC is critical for optimal efficacy and minimal undesired side effects. AREACOVERED This review addresses the challenges associated with the delivery of siRNA toHCC, discussing and focusing on delivery systems based on lipid and polymeric nanoparticles in publications from the past five years. EXPERT OPINION Future nano particles will need to effectively cross the vessel wall, migrate through the extracellular matrix and finally cross the HCC cell membrane. This may be achieved by optimizing nanoparticle size, the equipment of nanoparticles withHCC targeting moieties and loading nanoparticles with siRNAs againstHCC-specific oncogenes.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | | | | | - Mario Grassi
- Department of Engineering and Architecture, Trieste University, Trieste, Italy
| | - Domenico Tierno
- Clinical Department of Medical, Surgical and Health Sciences, Cattinara University Hospital, Trieste Univer-sity, Trieste, Italy
| | - Alice Biasin
- Department of Engineering and Architecture, Trieste University, Trieste, Italy
- Clinical Department of Medical, Surgical and Health Sciences, Cattinara University Hospital, Trieste Univer-sity, Trieste, Italy
| | - Nhung Hai Truong
- Laboratory of Regenerative Biomedicine, University of Science-VNUHCM, Ho Chi MInh City, Vietnam
- Faculty of Biology and Biotechnology, Viet Nam National University, Ho Chi Minh City, Vietnam
| | - Thanh Dang Minh
- Laboratory of Regenerative Biomedicine, University of Science-VNUHCM, Ho Chi MInh City, Vietnam
- Faculty of Biology and Biotechnology, Viet Nam National University, Ho Chi Minh City, Vietnam
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| | - Giorgia Pastorin
- Pharmacy Department, National University of Singapore, Singapore
| | - Federica Tonon
- Clinical Department of Medical, Surgical and Health Sciences, Cattinara University Hospital, Trieste Univer-sity, Trieste, Italy
| | - Gabriele Grassi
- Clinical Department of Medical, Surgical and Health Sciences, Cattinara University Hospital, Trieste Univer-sity, Trieste, Italy
| |
Collapse
|
209
|
Coskun A, Savas IN, Can O, Lippi G. From population-based to personalized laboratory medicine: continuous monitoring of individual laboratory data with wearable biosensors. Crit Rev Clin Lab Sci 2025; 62:198-227. [PMID: 39893518 DOI: 10.1080/10408363.2025.2453152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/28/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025]
Abstract
Monitoring individuals' laboratory data is essential for assessing their health status, evaluating the effectiveness of treatments, predicting disease prognosis and detecting subclinical conditions. Currently, monitoring is performed intermittently, measuring serum, plasma, whole blood, urine and occasionally other body fluids at predefined time intervals. The ideal monitoring approach entails continuous measurement of concentration and activity of biomolecules in all body fluids, including solid tissues. This can be achieved through the use of biosensors strategically placed at various locations on the human body where measurements are required for monitoring. High-tech wearable biosensors provide an ideal, noninvasive, and esthetically pleasing solution for monitoring individuals' laboratory data. However, despite significant advances in wearable biosensor technology, the measurement capacities and the number of different analytes that are continuously monitored in patients are not yet at the desired level. In this review, we conducted a literature search and examined: (i) an overview of the background of monitoring for personalized laboratory medicine, (ii) the body fluids and analytes used for monitoring individuals, (iii) the different types of biosensors and methods used for measuring the concentration and activity of biomolecules, and (iv) the statistical algorithms used for personalized data analysis and interpretation in monitoring and evaluation.
Collapse
Affiliation(s)
- Abdurrahman Coskun
- Department of Medical Biochemistry, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Irem Nur Savas
- Department of Medical Biochemistry, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ozge Can
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Giuseppe Lippi
- Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
210
|
You Q, Wang K, Zhao Z, Zhou H, Lan Z, Liang H, Deng R, Li W, Shen S, Wang R, Zhang K, Zheng D, Sun J. Reduction of Bacteroides fragilis in Gut Microbiome of Chronic Hepatitis B Patients Promotes Liver Injury. J Med Virol 2025; 97:e70395. [PMID: 40392071 DOI: 10.1002/jmv.70395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/03/2025] [Accepted: 05/04/2025] [Indexed: 05/22/2025]
Abstract
In chronic hepatitis B (CHB) patients under antiviral treatment, liver injury, as evidenced by elevated alanine transaminase (ALT), is associated with unfavorable outcomes and needs effective treatment. The interaction between gut microbiota and liver injury in CHB patients remains unclear. Using a case-control design, 28 cases with elevated ALT and 28 matched controls with normal ALT were randomly selected from CHB patients with viral control. Clinical characteristics were comparable between groups. Metagenomic sequencing revealed that Bacteroides fragilis was decreased in cases and exhibited the greatest disparity between cases and controls. Mice colonized by gut microbiota from cases exhibited more severe liver damage in both LPS-induced and MCD diet-induced liver injury models, and had a lower abundance of B. fragilis compared to mice colonized by gut microbiota from controls. Oral gavage of B. fragilis improved both LPS-induced and MCD diet-induced liver injury. Metabolomics analysis revealed that the levels of 7-Ketolithocholic acid (7-Keto-LCA) were positively correlated with B. fragilis and significantly increased in the cultural supernatant of B. fragilis. Consistently, 7-Keto-LCA exerted protective effects against both LPS-induced and MCD diet-induced liver damage. Targeting gut microbiota might be a promising therapeutic treatment for alleviation residual liver inflammation in CHB patients with viral control.
Collapse
Affiliation(s)
- Qiuhong You
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kaifeng Wang
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhou Zhao
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Heqi Zhou
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhixian Lan
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongyan Liang
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rui Deng
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Wanying Li
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sheng Shen
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Routing Wang
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kaikai Zhang
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dekai Zheng
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Sun
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
211
|
Wu J, Chen ZH, Xie Y, Fan Y. Advances in Lanthanide-Based NIR-IIb Probes for In Vivo Biomedical Imaging. SMALL METHODS 2025; 9:e2401462. [PMID: 39520332 DOI: 10.1002/smtd.202401462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/11/2024] [Indexed: 11/16/2024]
Abstract
The past decades have witnessed the significant development and practical interest of in vivo biomedical imaging technologies and optical materials in the second-near infrared (NIR-II, 1000-1700 nm) window. Imaging with the extended emission wavelength toward the long-wavelength end (NIR-IIb, 1500-1700 nm) further offers micrometer imaging resolution and centimeter tissue penetration depth by taking advantage of the much-reduced photon scattering and near-zero tissue autofluorescence background, which have become a very hot research area. This review focuses on the recent advances in the development of lanthanide-based NIR-IIb probes for in vivo biomedical applications. The progress including ratiometric imaging, multiplexed imaging for wide-field and microscopy, lifetime multiplexing and sensing, persistent luminescence, and multimodal imaging is summarized. Challenges and future directions concerning the investigation of the photophysical and photochemical properties of NIR-IIb probes, the selection of near-infrared cameras as well as the potential extension of the NIR-IIb imaging sub-window are pointed out. This review will inspire readers who have a strong interest in developing optical imaging technology and long-wavelength fluorescence probes for high-contrast in vivo biomedical applications.
Collapse
Affiliation(s)
- Jiaxin Wu
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, 200433, P. R. China
| | - Zi-Han Chen
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, 200433, P. R. China
| | - Yang Xie
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Yong Fan
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, 200433, P. R. China
| |
Collapse
|
212
|
Zhao Y, Wang H. Artificial intelligence-driven circRNA vaccine development: multimodal collaborative optimization and a new paradigm for biomedical applications. Brief Bioinform 2025; 26:bbaf263. [PMID: 40483546 PMCID: PMC12145227 DOI: 10.1093/bib/bbaf263] [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: 03/02/2025] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 06/11/2025] Open
Abstract
Circular RNA (circRNA) vaccines have emerged as a groundbreaking innovation in infectious disease prevention and cancer immunotherapy, offering superior stability and reduced immunogenicity compared to conventional linear messenger RNA (mRNA) vaccines. While linear mRNA vaccines are prone to degradation and can trigger strong innate immune responses, covalently closed circRNA vaccines leverage their unique circular structure to enhance molecular stability and minimize innate immune activation, positioning them as a next-generation platform for vaccine development. Artificial intelligence (AI) is revolutionizing circRNA vaccine design and optimization. Deep learning models, such as convolutional neural networks (CNNs) and Transformers, integrate multi-omics data to refine antigen prediction, RNA secondary structure modeling, and lipid nanoparticle delivery system formulation, surpassing traditional bioinformatics approaches in both accuracy and efficiency. While AI-driven bioinformatics enhances antigen screening and delivery system modeling, generative AI accelerates literature synthesis and experimental planning-though the risk of fabricated references and limited biological interpretability hinders its reliability. Despite these advancements, challenges such as the "black-box" nature of AI algorithms, unreliable literature retrieval, and insufficient integration of biological mechanisms underscore the necessity for a hybrid "AI-traditional-experimental" paradigm. This approach integrates explainable AI frameworks, multi-omics validation, and ethical oversight to ensure clinical translatability. Future research should prioritize mechanism-driven AI models, real-time experimental feedback, and rigorous ethical standards to fully unlock the potential of circRNA vaccines in precision oncology and global health.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Hematology, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an, Shaanxi 710061, P.R. China
| | - Huaiyu Wang
- Department of Hematology, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an, Shaanxi 710061, P.R. China
| |
Collapse
|
213
|
Kaur G, Ahmad A, Singh I. Recent progress in nanomaterial-based electrochemical biosensors for hydrogen peroxide detection & their biological applications. Talanta 2025; 286:127447. [PMID: 39753076 DOI: 10.1016/j.talanta.2024.127447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 03/03/2025]
Abstract
The electrochemical biosensor has brought a paradigm shift in the field of sensing due to its fast response and easy operability. The performance of electrochemical sensors can be modified by coupling them with various metal oxides, nanomaterials, and nanocomposites. Hydrogen peroxide is a short-lived reactive oxygen species that plays a crucial role in various physiological and biological processes. Therefore, its monitoring is of paramount importance. With this, the research fraternity has developed various nanomaterial-based superlative sensors that have enhanced the sensing performance towards H2O2 in terms of sensitivity, detection limit, and linear range. The integration of nanocomposite materials has allowed for the synergistic combination of different components, leading to improved sensor stability, selectivity, and detection limits. The precious metal alloys, metal oxides, semiconductor nanomaterials, carbon cloth, multi-walled carbon nanotubes, graphene oxide, and nanoparticles demonstrate effective catalytic performance for detecting H2O2 electrochemically. These advanced materials possess extraordinary properties and structures, rendering them highly advantageous for diverse applications. These biosensors aid in monitoring H2O2 levels secreted by MCF-7, HeLa cells, NIH-3T3, and A549 cells in real-time. Further, this type of biosensor identified alterations in H2O2 levels in the lungs, bronchoalveolar lavage fluid (BALF) of mice with pulmonary fibrosis, activated hepatic stellate cells, and the livers of mice with liver fibrosis. The current review highlights the recent advancements in compositions, morphology, limit of detection, sensitivity, biological applications, etc. properties of the electrochemical biosensors for H2O2 detection.
Collapse
Affiliation(s)
- Gurdeep Kaur
- School of Chemical Engineering and Physical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Aftab Ahmad
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Iqubal Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India.
| |
Collapse
|
214
|
Mao Q, Tian Y, Yu Q, Chen L, Zhang L, Tong Y, Yao W, Gao X, Tian H. Enhanced efficiency of MHC class II tumor neoantigen vaccines with a novel CD4 + T-cell helper epitope. J Pharmacol Exp Ther 2025; 392:103570. [PMID: 40267770 DOI: 10.1016/j.jpet.2025.103570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 03/23/2025] [Indexed: 04/25/2025] Open
Abstract
Tumor neoantigens, defined as tumor-specific antigens arising from somatic mutations, have shown great potential as targets for cancer vaccines in clinical studies. However, the number of neoantigens capable of effectively activating immune responses is quite limited. Over the past few decades, tumor neoantigen vaccines based on MHC-I epitopes that activate CD8+ T cells have been extensively studied. However, growing evidence suggests that CD4+ T cells are important in cancer immunotherapy. In contrast to CD8+ T cells, the receptors on CD4+ T cells exhibit a wider range of antigen peptide-MHC recognition, which can detect more tumor mutation antigens. In our earlier studies, a nitrated CD4+ T-cell epitope (NitraTh) was constructed as a novel CD4+ T-cell epitope that can enhance the immunogenicity of multiple tumor antigens. Therefore, we designed vaccines targeting MHC-II neoantigen epitopes using the nitrated T-cell epitope containing immunogenic amino acids. We found that vaccines conjugated with NitraTh exhibited enhanced immunogenicity. Crucially, the NitraTh-modified MHC-II tumor neoantigen vaccines increased the proportion of CD4+ T cells that infiltrate tumors and the spleen, elevated the expression of several cytokines with antitumor effects and facilitated the transformation of CD4+ T cells into Th1 cells, thereby reducing tumor growth. Additionally, the nitrated epitope has been shown to transform naïve CD4+ T cells into effector memory cells, thus facilitating enduring antitumor actions. The strategy of combining nitrated epitopes with MHC-II neoantigen epitopes confirms the significance of CD4+ T-cell immunity in cancer and may provide a novel approach for cancer vaccine design. SIGNIFICANCE STATEMENT: This study presents a novel design paradigm for tumor vaccines-combining MHC-II epitopes with nitrated CD4+ T-cell epitopes. This approach promotes the differentiation of CD4+ T cells toward a Th1 phenotype and generates long-lasting effector memory CD4+ T cells. Under the enhanced effects of CD4+ T cells, the vaccines we designed achieved superior antitumor efficacy and improved the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Qiuli Mao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yahong Tian
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qiumin Yu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Lingxiao Chen
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Li Zhang
- Department of General Internal Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yue Tong
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenbing Yao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| | - Hong Tian
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
215
|
Clarke HA, Hawkinson TR, Shedlock CJ, Medina T, Ribas RA, Wu L, Liu Z, Ma X, Xia Y, Huang Y, He X, Chang JE, Young LEA, Juras JA, Buoncristiani MD, James AN, Rushin A, Merritt ME, Mestas A, Lamb JF, Manauis EC, Austin GL, Chen L, Singh PK, Bian J, Vander Kooi CW, Evers BM, Brainson CF, Allison DB, Gentry MS, Sun RC. Glycogen drives tumour initiation and progression in lung adenocarcinoma. Nat Metab 2025; 7:952-965. [PMID: 40069440 PMCID: PMC12116239 DOI: 10.1038/s42255-025-01243-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/12/2025] [Indexed: 03/17/2025]
Abstract
Lung adenocarcinoma (LUAD) is an aggressive cancer defined by oncogenic drivers and metabolic reprogramming. Here we leverage next-generation spatial screens to identify glycogen as a critical and previously underexplored oncogenic metabolite. High-throughput spatial analysis of human LUAD samples revealed that glycogen accumulation correlates with increased tumour grade and poor survival. Furthermore, we assessed the effect of increasing glycogen levels on LUAD via dietary intervention or via a genetic model. Approaches that increased glycogen levels provided compelling evidence that elevated glycogen substantially accelerates tumour progression, driving the formation of higher-grade tumours, while the genetic ablation of glycogen synthase effectively suppressed tumour growth. To further establish the connection between glycogen and cellular metabolism, we developed a multiplexed spatial technique to simultaneously assess glycogen and cellular metabolites, uncovering a direct relationship between glycogen levels and elevated central carbon metabolites essential for tumour growth. Our findings support the conclusion that glycogen accumulation drives LUAD cancer progression and provide a framework for integrating spatial metabolomics with translational models to uncover metabolic drivers of cancer.
Collapse
Affiliation(s)
- Harrison A Clarke
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Tara R Hawkinson
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Cameron J Shedlock
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Terrymar Medina
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Roberto A Ribas
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Lei Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Zizhen Liu
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Xin Ma
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Biostatistics College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yi Xia
- Department of Biostatistics College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yu Huang
- Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, USA
- Regenstrief Institute, Indianapolis, IN, USA
- Department of Biostatistics and Health Data Science, School of Medicine, Indianapolis, IN, USA
| | - Xing He
- Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, USA
- Regenstrief Institute, Indianapolis, IN, USA
- Department of Biostatistics and Health Data Science, School of Medicine, Indianapolis, IN, USA
| | - Josephine E Chang
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Lyndsay E A Young
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Jelena A Juras
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | | | - Alexis N James
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Anna Rushin
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Matthew E Merritt
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Annette Mestas
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jessica F Lamb
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Elena C Manauis
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Grant L Austin
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Li Chen
- Department of Biostatistics College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Pankaj K Singh
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Jiang Bian
- Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, USA
- Regenstrief Institute, Indianapolis, IN, USA
- Department of Biostatistics and Health Data Science, School of Medicine, Indianapolis, IN, USA
| | - Craig W Vander Kooi
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - B Mark Evers
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Christine F Brainson
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Derek B Allison
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Matthew S Gentry
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA.
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA.
| | - Ramon C Sun
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA.
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA.
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
216
|
Wang C, Shi Y, Zhang D, Sun Y, Xie J, Wu B, Zhang C, Liu X. Generalization of neoantigen-based tumor vaccine by delivering peptide-MHC complex via oncolytic virus. EMBO Mol Med 2025; 17:1118-1152. [PMID: 40195559 DOI: 10.1038/s44321-025-00225-3] [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: 08/22/2024] [Revised: 03/06/2025] [Accepted: 03/14/2025] [Indexed: 04/09/2025] Open
Abstract
Neoantigen vaccine is a promising breakthrough in tumor immunotherapy. However, the application of this highly personalized strategy in the treatment of solid tumors is hindered by several obstacles, including very costly and time-consuming preparation steps, uncertainty in prediction algorithms and tumor heterogeneity. Universalization of neoantigen vaccine is an ideal yet currently unattainable solution to such limitations. To overcome these limitations, we engineered oncolytic viruses co-expressing neoantigens and neoantigen-binding major histocompatibility complex (MHC) molecules to force ectopic delivery of peptide-MHC ligands to T cell receptors (TCRs), enabling specific targeting by neoantigen vaccine-primed host immunity. When integrated with neoantigen vaccination, the engineered viruses exhibited potent cytolytic activity in a variety of tumor models irrespective of the neoantigen expression profiles, eliciting robust systemic antitumor immunity to reject tumor rechallenge and inhibit abscopal tumor growth with a favorable safety profile. Thus, this study provides a powerful approach to enhance the universality and efficacy of neoantigen vaccines, meeting the urgent need for universal neoantigen vaccines in the clinic to facilitate the further development of tumor immunotherapy.
Collapse
Affiliation(s)
- Chenyi Wang
- College of Chemical Engineering, Fuzhou University, Fuzhou, 350108, P. R. China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Yingjun Shi
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Yupeng Sun
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Junjie Xie
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Bingchen Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Cuilin Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China.
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, P. R. China.
| | - Xiaolong Liu
- College of Chemical Engineering, Fuzhou University, Fuzhou, 350108, P. R. China.
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China.
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, P. R. China.
| |
Collapse
|
217
|
Aghaei‐Zarch SM, Esmaeili A, Bagheri‐Mohammadi S. A Comprehensive Review on LncRNAs/miRNAs-DNMT1 Axis in Human Cancer: Mechanistic and Clinical Application. J Cell Mol Med 2025; 29:e70604. [PMID: 40387409 PMCID: PMC12087000 DOI: 10.1111/jcmm.70604] [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: 08/19/2024] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025] Open
Abstract
Cancer constitutes a significant public health concern, and addressing the challenge of cancer holds paramount importance and requires immediate attention. Epigenetic alterations, encompassing DNA methylation, have emerged as pivotal contributors to the development of diverse cancer types. These modifications exert their influence by modulating chromatin structure, gene expression patterns and other nuclear processes, thereby influencing cancer pathogenesis. Over the last two decades, an increasing body of evidence has established the involvement of DNA methyltransferase 1 (DNMT1) in various aspects of cancer development, including tumorigenesis, aggressiveness and treatment response. Furthermore, non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), are increasingly recognised as significant modulators in diverse biological processes, encompassing metastasis, apoptosis, cell proliferation and differentiation. Several recent studies have elucidated the intricate relationship between epigenetic machinery, specifically DNMT1, and the expression of ncRNAs in the context of cancer. In this review, we provide a comprehensive overview of the interaction between DNMT1 and ncRNAs in cancer pathogenesis. Furthermore, we discuss the important role of the ncRNAs-DNMT1 axis in cancer stem cells and cancer therapy resistance as critical issues in cancer therapy. Finally, we demonstrate that herbal medicine and synthetic RNA molecules regulate DNMT1 activity and hold great promise in cancer treatment.
Collapse
Affiliation(s)
- Seyed Mohsen Aghaei‐Zarch
- Department of Medical Genetics, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Ali Esmaeili
- Student Research Committee, Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Saeid Bagheri‐Mohammadi
- Department of Paramedicine, Amol School of Paramedical SciencesMazandaran University of Medical SciencesSariIran
- Immunogenetic Research CenterMazandaran University of Medical SciencesSariIran
| |
Collapse
|
218
|
Xie M, Jiang C, Zhang C, Wu Y, Zhang X, Yao R, Han C, Dai Y, Xu K, Zheng S. Tumor microenvironment triggered iron-based metal organic frameworks for magnetic resonance imaging and photodynamic-enhanced ferroptosis therapy. J Colloid Interface Sci 2025; 685:382-395. [PMID: 39855085 DOI: 10.1016/j.jcis.2025.01.119] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Photodynamic therapy (PDT) primarily relies on the generation of reactive oxygen species (ROS) to eliminate tumor cells. However, the elevated levels of glutathione (GSH) within tumor cells can limit the efficacy of PDT, posing a challenge to achieve complete tumor eradication. Herein, a porous iron-based metal-organic frameworks (PEG-Fe-MOFs) nanoplatform was developed for the combined application of PDT and ferroptosis in cancer treatment. The coordination between tetrakis (4-carboxyphenyl) porphyrin (TCPP) and ferric (Fe3+) enabled PEG-modified Fe-MOFs (PEG-Fe-MOFs) to deliver excellent T1-weighted magnetic resonance (MR) imaging performance in physiological environments. Within the tumor microenvironment (TME), PEG-Fe-MOFs gradually degraded to release TCPP, which could be utilized for fluorescence imaging. Moreover, Fe2+ enhanced intracellular ROS levels via the Fenton reaction, generating hydroxyl radicals that further amplified ROS production. This synergistic effect comprising increased ROS levels and GSH depletion augmented the efficacy of PDT while simultaneously inducing robust ferroptosis in tumor cells, thereby maximizing therapeutic outcomes. Both in vitro and in vivo experiments have demonstrated the superior T1 weighted MR and fluorescence imaging capabilities of PEG-Fe-MOFs, along with its potent synergistic therapeutic effects on tumors. These results highlighted the potential of this nanoplatform for combining PDT and ferroptosis in cancer treatment.
Collapse
Affiliation(s)
- Manman Xie
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Canran Jiang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Cong Zhang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Yun Wu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Xiuli Zhang
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Ruosi Yao
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Cuiping Han
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China.
| | - Yue Dai
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China.
| | - Kai Xu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China; Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China.
| | - Shaohui Zheng
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China; Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
219
|
Cheng Z, Ren Y, Wang X, Zhang Y, Hua Y, Zhao H, Lu H. A novel prognostic framework for HBV-infected hepatocellular carcinoma: insights from ferroptosis and iron metabolism proteomics. Brief Bioinform 2025; 26:bbaf216. [PMID: 40381315 PMCID: PMC12085197 DOI: 10.1093/bib/bbaf216] [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: 12/02/2024] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 05/20/2025] Open
Abstract
Effective classification methods and prognostic models enable more accurate classification and treatment of hepatocellular carcinoma (HCC) patients. However, the weak correlation between RNA and protein data has limited the clinical utility of previous RNA-based prognostic models for HCC. In this work, we constructed a novel prognostic framework for HCC patients using seven differentially expressed proteins associated with ferroptosis and iron metabolism. Furthermore, this prognostic model robustly classifies HCC patients into three clinically relevant risk groups. Significant differences in overall survival, age, tumor differentiation, microvascular invasion, distant metastasis, and alpha-fetoprotein levels were observed among the risk groups. Based on the prognostic model and known biological pathways, we explored the potential mechanisms underlying the inconsistent differential expression patterns of FTH1 (Ferritin heavy chain 1) mRNA and protein. Our findings demonstrated that tumor tissues in HCC patients promote liver cancer progression by downregulating FTH1 protein expression, rather than upregulating FTH1 mRNA expression, ultimately leading to poor prognosis. Subsequently, based on risk score and tumor size, we developed a nomogram for predicting the prognosis of HCC patients, which demonstrated superior predictive performance in both the training and validation cohorts (C-index: 0.774; AUC for 1-5 years: 0.783-0.964). Additionally, our findings demonstrated that the adverse prognosis of high-risk HCC patients was closely correlated with ferroptosis in liver cancer tissues, alterations in iron metabolism, and changes in the tumor immune microenvironment. In conclusion, our prognostic model and predictive nomogram offer novel insights and tools for the effective classification of HCC patients, potentially enhancing clinical decision-making and outcomes.
Collapse
Affiliation(s)
- Zhiwei Cheng
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- Department of Orthopedic Oncology, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Hongkou District, Shanghai 200080, China
- SJTU-Yale Joint Center of Biostatistics and Data Science, National Center for Translational Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Yongyong Ren
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- Institute of Bioinformatics, Shanghai Academy of Experimental Medicine, 528 Hongshan Road, Pudong New District, Shanghai 200126, China
| | - Xinbo Wang
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- SJTU-Yale Joint Center of Biostatistics and Data Science, National Center for Translational Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Yuening Zhang
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- SJTU-Yale Joint Center of Biostatistics and Data Science, National Center for Translational Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Yingqi Hua
- Department of Orthopedic Oncology, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Hongkou District, Shanghai 200080, China
| | - Hongyu Zhao
- Department of Biostatistics, Yale University, 300 George Street, New Haven, CT 06511, United States
| | - Hui Lu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- SJTU-Yale Joint Center of Biostatistics and Data Science, National Center for Translational Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- Institute of Bioinformatics, Shanghai Academy of Experimental Medicine, 528 Hongshan Road, Pudong New District, Shanghai 200126, China
- Shanghai Engineering Research Center for Big Data in Pediatric Precision Medicine, Center for Biomedical Informatics, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, 1400 Beijing West Road, Jing'an District, Shanghai 200040, China
| |
Collapse
|
220
|
Lan Z, Zheng J, Zhang X, Zhang J, Chen Z, Chen Y, Yan S, Peng Y, Yu X. Enhancing prediction of parenchymal hemorrhage type 2 after endovascular treatment in acute ischemic stroke using dual-phase CTA. Eur J Radiol 2025; 186:112027. [PMID: 40043546 DOI: 10.1016/j.ejrad.2025.112027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/09/2025] [Accepted: 02/27/2025] [Indexed: 04/07/2025]
Abstract
OBJECTIVE To evaluate the effectiveness of dual-phase CT angiography (CTA) in predicting parenchymal hemorrhage type 2 (PH2) following endovascular thrombectomy (EVT) in patients with acute ischemic stroke (AIS). METHODS A retrospective analysis was conducted across two centers, including 232 AIS patients who underwent EVT. Three predictive models were developed: a clinical model (Model C), a clinical model incorporating single-phase CTA data (Model CS), and a clinical model incorporating dual-phase CTA data (Model CD). The performance of these models in predicting PH2 occurrence post-EVT was assessed and compared. RESULTS The model incorporating dual-phase CTA data (Model CD) demonstrated superior predictive performance, with higher area under the curve (AUC) values in both training and validation datasets, compared to Models C and CS. Calibration and decision curve analyses further confirmed the enhanced accuracy and clinical utility of Model CD. CONCLUSION The findings indicate that dual-phase CTA provides a more accurate assessment of collateral circulation compared to single-phase CTA, thereby improving the prediction of PH2 after EVT. This enhanced predictive capability can assist clinicians in making more informed decisions regarding AIS management.
Collapse
Affiliation(s)
- Zhihong Lan
- Department of Radiology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, People's Republic of China
| | - Jiakai Zheng
- Department of Radiology, Shenshan Medical Center, Memorial Hospital of Sun Yat-Sen University, Shanwei, People's Republic of China
| | - Xiaoling Zhang
- Department of Radiology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, People's Republic of China
| | - Jiawei Zhang
- Department of Radiology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, People's Republic of China
| | - Zhiyan Chen
- Department of Radiology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, People's Republic of China
| | - Yafang Chen
- Department of Radiology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, People's Republic of China
| | - Shuyue Yan
- Department of Radiology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, People's Republic of China
| | - Yongjun Peng
- Department of Radiology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, People's Republic of China.
| | - Xiangrong Yu
- Department of Radiology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, People's Republic of China.
| |
Collapse
|
221
|
Liu J, Cheng P, Xu C, Pu K. Molecular probes for in vivo optical imaging of immune cells. Nat Biomed Eng 2025; 9:618-637. [PMID: 39984703 DOI: 10.1038/s41551-024-01275-7] [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: 05/23/2023] [Accepted: 09/23/2024] [Indexed: 02/23/2025]
Abstract
Advancing the understanding of the various roles and components of the immune system requires sophisticated methods and technology for the detection of immune cells in their natural states. Recent advancements in the development of molecular probes for optical imaging have paved the way for non-invasive visualization and real-time monitoring of immune responses and functions. Here we discuss recent progress in the development of molecular probes for the selective imaging of specific immune cells. We emphasize the design principles of the probes and their comparative performance when using various optical modalities across disease contexts. We highlight molecular probes for imaging tumour-infiltrating immune cells, and their applications in drug screening and in the prediction of therapeutic outcomes of cancer immunotherapies. We also discuss the use of these probes in visualizing immune cells in atherosclerosis, lung inflammation, allograft rejection and other immune-related conditions, and the translational opportunities and challenges of using optical molecular probes for further understanding of the immune system and disease diagnosis and prognosis.
Collapse
Affiliation(s)
- Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
222
|
Zhang X, Wu M, Shi H, Kim S, Lu S, Wang P, Qin J. Amplification-Free Electrochemiluminescent Biosensor for Ultrasensitive Detection of Fusobacterium nucleatum Using Tetrahedral DNA-Based CRISPR/Cas12a. CYBORG AND BIONIC SYSTEMS 2025; 6:0266. [PMID: 40313468 PMCID: PMC12044220 DOI: 10.34133/cbsystems.0266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Fusobacterium nucleatum, a bacterium linked to colorectal cancer, possesses a specific gene called fadA that serves as an early diagnostic biomarker. The CRISPR/Cas12a system has demonstrated marked potential for nucleic acid detection due to its satisfactory selectivity and trans-cleavage ability. However, most CRISPR/Cas-based sensors suffer from problems such as probe entanglement or local aggregation, reducing the Cas enzyme efficiency. In this study, an amplification-free biosensing platform for ultrasensitive detection of F. nucleatum was developed by integrating the highly specific CRISPR/AsCas12a with an improved electrochemiluminescence (ECL) biosensor. Different from the conventional 1- or 2-dimensional probes, the platform was constructed by tetrahedral DNA nanostructure (TDN) probes conjugated with quenchers and coralliform gold (CFAu) functionalized with luminescent agents. The TDN serves as an exceptional scaffold to modulate the recognition unit, substantially enhancing the recognition and cleavage efficiency of AsCas12a toward the probes. Furthermore, the high surface area of CFAu provides extensive landing sites for the luminescent agents, thereby improving the detection sensitivity. The prepared ECL biosensor exhibited a wider linear range (10 fM to 100 nM) and was capable of detecting F. nucleatum down to 1 colony-forming unit/ml. Additionally, the high mismatch sensitivity of AsCas12a to protospacer adjacent motifs and nearby areas provides a strategy for distinguishing mutant from wild-type sequences. Finally, by designing CRISPR RNA (crRNA), this diagnostic method can also be easily modified to detect other bacteria or biomarkers for the early diagnosis of various diseases.
Collapse
Affiliation(s)
- Xindan Zhang
- School of Chemistry and Chemical Engineering,
Beijing Institute of TechnologyZhengzhou Academy of Intelligent Technology, Beijing Institute of Technology, Beijing 100081, China
- Tongji UniversityCancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Minkang Wu
- Tongji UniversityCancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Haoran Shi
- School of Bioengineering,
Dalian University of Technology, Dalian City, Liaoning 116024, China
| | - Soochan Kim
- School of Chemical Engineering,
Sungkyunkwan University, Seoul, South Korea
| | - Shixiang Lu
- School of Chemistry and Chemical Engineering,
Beijing Institute of TechnologyZhengzhou Academy of Intelligent Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Ping Wang
- Tongji UniversityCancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jieling Qin
- School of Chemistry and Chemical Engineering,
Beijing Institute of TechnologyZhengzhou Academy of Intelligent Technology, Beijing Institute of Technology, Beijing 100081, China
- Tongji UniversityCancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
223
|
Zhang M, Ji Y, Liu M, Dai Y, Zhang H, Tong S, Cai Y, Liu M, Qu N. Nano-delivery of STING agonists: Unraveling the potential of immunotherapy. Acta Biomater 2025; 197:104-120. [PMID: 40164370 DOI: 10.1016/j.actbio.2025.03.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
The cyclic GMP-AMP synthetase-interferon gene stimulator (cGAS-STING) pathway possesses tremendous potential in immune responses, viral defense, and anti-tumor treatment. Currently, an increasing number of nanocarriers are being engineered to convey STING agonists, with the goal of booSTING the conveying capacity of cGAS-STING agonists and augment the therapeutic potency of STING agonists. In this review, we explore the mechanisms of cGAS-STING activators, the application of different nanocarriers in the STING pathway, and the application of nanocarriers in anti-tumor therapy, antiviral therapy and autoimmune diseases. Additionally, we also discuss the adverse effects of STING pathway activation and the challenges encountered in nano delivery, we hope that future research will delve into the development of new nanocarriers and the clinical translation of nanocarriers in STING-mediated immunotherapy. STATEMENT OF SIGNIFICANCE: The cyclic GMP-AMP synthetase-interferon gene stimulator (cGAS-STING) pathway possesses tremendous potential in immune responses, viral defense, and anti-tumor treatment. In this review, we first explore the activation mechanism of cGAS-STING signal pathway and the diverse array of nanocarriers that have been employed in the context of the STING pathway, such as natural carrier, lipid nanoparticles, polymeric nanoparticles, and inorganic nanoparticles, highlighting their unique properties and the challenges they present in clinical applications. Furthermore, we discuss the research progress regarding nanocarriers in STING-mediated immunotherapy, such as the application of nanocarriers in anti-tumor therapy, antiviral therapy and autoimmune diseases therapy. Finally, the side effects of STING pathway activation and the issues encountered in nano delivery will be discussed, hoping that future research will delve into the development of new nanocarriers and the clinical translation of nanocarriers in STING-mediated immunotherapy.
Collapse
Affiliation(s)
- Meng Zhang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Yating Ji
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Mingxia Liu
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Yixin Dai
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Hongxia Zhang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Shiyu Tong
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Yuqing Cai
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Mengjiao Liu
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Na Qu
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China.
| |
Collapse
|
224
|
Chen L, Li Y, Zhang N, Chen T, Li F, Han J, Wang Z, Kannan PR, Sun Z, Fu F, Cheng L, Lu J, Kong X. Injectable dual-cross-linked microalgae-silk gel ameliorates diabetic wound healing by promoting oxygenation and ROS clearance and lessening inflammation. Int J Biol Macromol 2025; 309:142897. [PMID: 40203918 DOI: 10.1016/j.ijbiomac.2025.142897] [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: 01/13/2025] [Revised: 04/01/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Hypoxia, excessive reactive oxygen species (ROS), and an impaired inflammatory microenvironment are key barriers to diabetic wound healing, collectively hindering cell migration, proliferation, and neovascularization, ultimately leading to failure in the healing process. Therefore, developing an effective therapeutic strategy capable of simultaneously addressing these challenges remains a critical clinical need. In this study, we developed CeS-Gel, an advanced hydrogel dressing integrating live microalgae and CeO₂ nanoparticles within a dual-crosslinked silk hydrogel network. By harnessing photosynthesis, CeS-Gel provided a continuous and reliable oxygen supply, significantly enhancing cell migration and proliferation. Additionally, CeS-Gel exhibited potent ROS-scavenging properties, effectively mitigating oxidative stress-induced cellular damage while directly promoting M2 macrophage polarization, thereby modulating the inflammatory response. In vivo experiments demonstrated that CeS-Gel markedly accelerated wound healing in diabetic mice, achieving a 93.2 % wound closure rate. Furthermore, CeS-Gel effectively alleviated hypoxia, promoted neovascularization, and exhibited anti-inflammatory and immunoregulatory effects. This living microalgae-silk gel represents a promising approach for improving chronic diabetic wound healing with great potential for clinical application.
Collapse
Affiliation(s)
- Liuting Chen
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Yao Li
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Na Zhang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Tianshuang Chen
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Feiyan Li
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Jiayi Han
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Zihang Wang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Perumal Ramesh Kannan
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Zeyue Sun
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Feiya Fu
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Ling Cheng
- Luoxi Medical Technology (Hangzhou) Co., Ltd., Hangzhou 310018, China
| | - Jiaju Lu
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou 310018, China.
| | - Xiangdong Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China; International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou 310018, China.
| |
Collapse
|
225
|
Ding S, Hao Y, Qi Y, Wei H, Zhang J, Li H. Molecular mechanism of tumor-infiltrating immune cells regulating endometrial carcinoma. Genes Dis 2025; 12:101442. [PMID: 40083326 PMCID: PMC11904505 DOI: 10.1016/j.gendis.2024.101442] [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: 11/09/2023] [Revised: 07/14/2024] [Accepted: 08/14/2024] [Indexed: 03/16/2025] Open
Abstract
Endometrial carcinoma (EC) is a prevalent gynecological cancer, and its interaction with the immune system is pivotal in cancer progression. This comprehensive review explores the molecular mechanisms involved in the regulation of EC by tumor-infiltrating immune cells. This review discusses the composition and functions of various immune cell types within the tumor microenvironment, including T cells, B cells, macrophages, and natural killer cells, and elucidates their specific roles in cancer control. It also delves into the immune evasion strategies employed by EC cells, with a specific focus on immune checkpoint pathways and their influence on tumor development. Signaling pathways, cytokines, and chemokines mediating immune responses within the tumor microenvironment are also detailed. Furthermore, clinical implications and therapeutic strategies, such as immunotherapies, are also reviewed, and relevant clinical trials are discussed. Additionally, this review discusses the existing gaps in our knowledge, suggests potential avenues for future research, and emphasizes the significance of understanding these mechanisms for enhanced EC treatment. This review provides an exhaustive overview of the current knowledge, supporting the ongoing quest for more effective therapeutic interventions on EC.
Collapse
Affiliation(s)
- Silu Ding
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 117004, China
| | - Yingying Hao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 117004, China
| | - Yue Qi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 117004, China
| | - Heng Wei
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 117004, China
| | - Jin Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 117004, China
| | - Hui Li
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, Liaoning 117004, China
| |
Collapse
|
226
|
Vasanth A, Ashok A, Do TN, Phan HP. Advancements in flexible porous Nanoarchitectonic materials for biosensing applications. Adv Colloid Interface Sci 2025; 339:103439. [PMID: 39978155 DOI: 10.1016/j.cis.2025.103439] [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: 11/02/2024] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 02/22/2025]
Abstract
The development of nanoporous materials has gained significant attention due to their unique structural properties and multimodalities, which are highly relevant for advanced sensing technologies. The capability to directly grow nanoporous materials on flexible substrates or indirectly integrate them into soft templates through mixing and dispersion opens exciting opportunities for a new class of flexible and stretchable electronics for personalized healthcare applications. This review paper provides a snapshot of recent advancements in flexible nanoporous materials and their applications, emphasizing biological and biomedical sensors. The review highlights the material of choice for flexible and stretchable substrates and effective approaches to synthesize and integrate nanoporous architectures onto soft polymers. Applications from wearable sweat sensors, mechanical sensors for electronic skins, implantable bioelectronics, and gas sensing are also presented. The paper concludes with current challenges and future perspectives within this highly active research paradigm.
Collapse
Affiliation(s)
- Arya Vasanth
- School of Mechanical and Manufacturing Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia; Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Aditya Ashok
- School of Mechanical and Manufacturing Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia; Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia.
| | - Thanh Nho Do
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia; Tyree Foundation Institute of Health Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Hoang-Phuong Phan
- School of Mechanical and Manufacturing Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia; Tyree Foundation Institute of Health Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia.
| |
Collapse
|
227
|
Liu X, Yang D, Liu B, Tang D. Progress in Signal Amplification and Microstructure Manufacturing for Photoelectrochemical Sensing. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2025; 18:383-405. [PMID: 40372815 DOI: 10.1146/annurev-anchem-070524-093940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
Photoelectrochemical (PEC) sensing based on chemical or biological recognition has received a tremendous amount of attention in recent years, providing analytical chemists a plethora of opportunities. However, emerging techniques and unknown processes in this field remain unexplored. We summarize the recently reported PEC sensing methods. First, we briefly describe the basic principles and technical characteristics of PEC sensing. Next, we highlight the application of various materials, nucleic acids, and other strategies for amplifying PEC signals. Finally, we discuss the current state of knowledge regarding the realization of miniaturized equipment during PEC sensor manufacturing. Summarizing the technological advances and research breakthroughs in PEC sensing over time can help increase the quality of follow-up research.
Collapse
Affiliation(s)
- Xu Liu
- Guizhou Engineering Laboratory for Synthetic Drugs (Ministry of Education of Guizhou Province), College of Pharmacy, Guizhou University, Guiyang, China;
| | - Di Yang
- Guizhou Engineering Laboratory for Synthetic Drugs (Ministry of Education of Guizhou Province), College of Pharmacy, Guizhou University, Guiyang, China;
| | - Bingqian Liu
- Guizhou Engineering Laboratory for Synthetic Drugs (Ministry of Education of Guizhou Province), College of Pharmacy, Guizhou University, Guiyang, China;
| | - Dianping Tang
- Key Laboratory of Analytical Science for Food Safety and Biology (Ministry of Education), Department of Chemistry, Fuzhou University, Fuzhou, China;
| |
Collapse
|
228
|
Attique I, Haider Z, Khan M, Hassan S, Soliman MM, Ibrahim WN, Anjum S. Reactive Oxygen Species: From Tumorigenesis to Therapeutic Strategies in Cancer. Cancer Med 2025; 14:e70947. [PMID: 40377005 DOI: 10.1002/cam4.70947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Reactive oxygen species (ROS), a class of highly reactive molecules, are closely linked to the pathogenesis of various cancers. While ROS primarily originate from normal cellular processes, external stimuli can also contribute to their production. Cancer cells typically exhibit elevated ROS levels due to disrupted redox homeostasis, characterized by an imbalance between antioxidant and oxidant species. ROS play a dual role in cancer biology: at moderate levels, they facilitate tumor progression by regulating oncogenes and tumor suppressor genes, inducing mutations, promoting proliferation, extracellular matrix remodeling, invasion, immune modulation, and angiogenesis. However, excessive ROS levels can cause cellular damage and initiate apoptosis, necroptosis, or ferroptosis. METHODS This review explores molecular targets involved in redox homeostasis dysregulation and examines the impact of ROS on the tumor microenvironment (TME). Literature from recent in vitro and in vivo studies was analyzed to assess how ROS modulation contributes to cancer development and therapy. RESULTS Findings indicate that ROS influence cancer progression through various pathways and cellular mechanisms. Targeting ROS synthesis or enhancing ROS accumulation in tumor cells has shown promising anticancer effects. These therapeutic strategies exhibit significant potential to impair tumor growth while also interacting with elements of the TME. CONCLUSION The ROS serve as both promoters and suppressors of cancer depending on their intracellular concentration. Their complex role offers valuable opportunities for targeted cancer therapies. While challenges remain in precisely modulating ROS for therapeutic benefit, they hold promise as synergistic agents alongside conventional treatments, opening new avenues in cancer management.
Collapse
Affiliation(s)
- Iqra Attique
- Department of Biotechnology, Kinnaird College for Women University, Lahore, Pakistan
| | - Zahra Haider
- Department of Biotechnology, Kinnaird College for Women University, Lahore, Pakistan
| | - Maha Khan
- Department of Biotechnology, Lahore College for Women University, Lahore, Pakistan
| | - Samina Hassan
- Department of Botany, Kinnaird College for Women University, Lahore, Pakistan
| | - Mohamed Mohamed Soliman
- Clinical Laboratory Sciences Department, Turabah University College, Taif University, Taif, Saudi Arabia
- Biochemistry Department, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Sumaira Anjum
- Department of Biotechnology, Kinnaird College for Women University, Lahore, Pakistan
| |
Collapse
|
229
|
Ahmadi Y, Yu Y, Cui Z, Huang WE, Andersson MI. Loop-Mediated Isothermal Amplification (LAMP) for the Diagnosis of Sexually Transmitted Infections: A Review. Microb Biotechnol 2025; 18:e70153. [PMID: 40317856 PMCID: PMC12047449 DOI: 10.1111/1751-7915.70153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 05/07/2025] Open
Abstract
Sexually transmitted infections (STIs) remain a significant public health concern. Given the asymptomatic nature of many STIs, diagnostic testing is critical for determining the appropriate treatment, enabling effective tracing and reducing the risk of further transmission. Nucleic acid amplification tests (NAATs) are the most sensitive and the most widely used in well-resourced settings. The majority of available NAATs are based on polymerase chain reaction (PCR), which requires highly trained personnel and costly equipment, making it impractical for resource-limited settings. Loop-mediated isothermal amplification (LAMP) has emerged as a simple, rapid, sensitive and low-cost alternative for pathogen detection, particularly well-suited for point-of-care tests (POCT). In this review, we evaluate LAMP assays reported in the literature for the detection of pathogens linked to the high incidence STIs prioritised by the World Health Organization (WHO) for POCT in 2023. These include Neisseria gonorrhoeae, Chlamydia trachomatis, Trichomonas vaginalis, T. pallidum subspecies pallidum, as well as other common STIs such as herpes simplex virus, hepatitis B virus and human immunodeficiency virus (HIV). For each LAMP assay, we identified and summarised the key elements such as the type and number of tested clinical specimens, chosen target gene, detection system, reference test and clinical outcomes. We highlight the advantages and limitations of these assays and discuss the gaps that should be addressed to improve their applicability for POCT.
Collapse
Affiliation(s)
- Yasaman Ahmadi
- Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Yejiong Yu
- Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Zhanfeng Cui
- Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Wei E. Huang
- Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Monique I. Andersson
- Department of MicrobiologyOxford University Hospitals NHS Foundation TrustOxfordUK
- Nuffield Division of Clinical Laboratory Science, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
230
|
Liu H, Wang D, Feng X, Liu L, Liu B, Zhu L, Sun J, Zuo X, Chen S, Liu J, Xian J, Zhang C, Yang W. Angelicin ameliorated ulcerative colitis through activating HDAC1-derived HIF-1α acetylation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156546. [PMID: 40058317 DOI: 10.1016/j.phymed.2025.156546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/12/2025] [Accepted: 02/15/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is a stubborn disease that occurs globally. SSP-TXYF is a traditional Chinese medicine (TCM) compound, which is widely used in the treatment of UC, but its picking and preparation is complicated. Therefore, this work analyzes the components of SSP-TXYF to find an natural compound that can treat UC, and verifies its efficacy and mechanism, aiming to explore new natural compound with the same efficacy as SSP-TXYF. METHODS AND RESULTS In this study, we used the quality markers (Q-marker) strategy to analyze the SSP-TXYF. We obtained that angelicin (Ang) is the main active ingredient. Combined with reductive dimethyl labeling and tandem orthogonal proteolysis-activity-based protein profiling (rdTOP-ABPP) and network pharmacology, we found that ERK1/2 and HDAC1 may be the binding proteins. Then, we demonstrated that Ang not only suppresses inflammation in lipopolysaccharide-induced IEC-6 but also effectively inhibits 2,4,6-trinitrobenzenesulfonic acid-induced UC in vivo. Increasing the concentration of propionate and butyrate, activating GPR43 and GPR109A receptors, increasing ERK1/2 activity, promoting Sp1 phosphorylation, thus competitively inhibiting the combination of HIF-1α and HDAC1, and finally increasing the acetylation of HIF-1α is the molecular signaling mechanism for Ang. CONCLUSIONS In conclusion, Ang can improve the symptoms of UC in rats and activate the GPR/ERK/SP1/HDAC1/HIF-1α pathway in colonic epithelial cells to repair the damaged intestinal mucosal barrier (IMB). The key pharmacodynamic mechanism of Ang is consistent with SSP-TXYF, and it is preliminarily believed that Ang can be used as a new natural medicine for treating UC.
Collapse
Affiliation(s)
- Haifan Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dunfang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xue Feng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingwei Sun
- Beijing University of Chinese Medicine, Beijing, China
| | - Xingbo Zuo
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Siyuan Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jianyao Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junying Xian
- Nanning Hospital of Traditional Chinese Medicine, Nanning, China
| | - Caijuan Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
231
|
Sasanipoor F, Zhang Z. Molybdenum Disulfide Nanocomposites for Cancer Diagnosis and Therapeutics: Biosensors, Bioimaging, and Phototherapy. Adv Healthc Mater 2025; 14:e2500655. [PMID: 40289409 DOI: 10.1002/adhm.202500655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/10/2025] [Indexed: 04/30/2025]
Abstract
Molybdenum disulfide (MoS₂) nanomaterials have attracted significant interest in cancer diagnosis and therapy due to their unique physicochemical properties. Due to its extensive surface area and adaptable structure, MoS₂ may engage with pharmaceuticals and biomolecules via covalent and non-covalent interactions. This versatility enhances the sensitivity of identifying specific biomarkers, colloidal stability, and tumor-targeting capabilities. In the near-infrared (NIR) spectrum, MoS₂ exhibits strong optical absorption and efficient photothermal conversion, making it suitable for NIR-driven phototherapy and regulated medication release. Functionalized MoS₂ nanocomposites react differently to the tumor microenvironment, which improves treatment effectiveness by increasing drug accumulation at cancer sites and decreasing off-target effects on healthy tissues. Recent developments in MoS₂-based nanocomposites for cancer detection and treatment are reviewed in this study, with particular attention paid to their uses in photothermal therapy, photodynamic therapy, biosensing, and bioimaging. Additionally, it looks at the difficulties and potential applications of MoS₂ nanocomposites in cancer.
Collapse
Affiliation(s)
- Fatemeh Sasanipoor
- Department of Chemistry, College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao, 266580, China
| | - Zhiqing Zhang
- Department of Chemistry, College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao, 266580, China
| |
Collapse
|
232
|
Cheng M, Liu J, Liang Y, Xu J, Ma L, Liang J. Tissue-Resident Memory T Cells in Tumor Immunity and Immunotherapy of Digestive System Tumors. Immunol Invest 2025; 54:435-456. [PMID: 39840686 DOI: 10.1080/08820139.2024.2447780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
BACKGROUND Tissue-resident memory T (TRM) cells possess unique abilities to migrate, establish themselves in tissues, and monitor peripheral tissues without circulating. They are crucial in providing long-lasting and local immune protection against surface infections. TRMs demonstrate distinct phenotypic and functional characteristics compared to central memory T (Tcm) cells and effector memory T (Tem) cells. METHODS We reviewed a large number of literature to explore the physiological and functional roles of tissue-resident memory T cells, as well as the link between TRM cells and the development and prognosis of digestive tract tumors. We also investigated the association between TRM cells, intestinal flora, and metabolites. RESULTS Recent studies have implicated TRMs in the immune response against tumors, making them a potential target for cancer therapy. However, research specifically focused on gastrointestinal tumors is limited. CONCLUSION This review aims to compile and assess the most recent data on the role of TRM cells in gastrointestinal tumor immunity. Additionally, it explores recent advancements in immunotherapy and investigates how TRMs may influence intestinal flora and metabolites.
Collapse
Affiliation(s)
- Min Cheng
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Jie Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Yue Liang
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of General Surgery (Breast Surgery), The First Affiliated Hospital of Shandong First Medical University (Shandong Provincial Qianfoshan Hospital), Jinan, China
| | - Jiamei Xu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Lin Ma
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| |
Collapse
|
233
|
Xu Y, Wang B, Huang Y, Liao J, Wu C, Zhou C, Kang Z, Jiang S, Wu B, Zhang D, Xu R, Liu X, Wang F. Targeting Antigen-Presenting Cells to Enhance the Tumor-Spleen Immunity Cycle through Liposome-Neoantigen Vaccine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500021. [PMID: 40125791 PMCID: PMC12097013 DOI: 10.1002/advs.202500021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 03/03/2025] [Indexed: 03/25/2025]
Abstract
Effective immune responses in both the spleen and the tumor microenvironment are crucial for cancer immunotherapy. However, delivery of neoantigen peptide vaccines to antigen-presenting cells (APCs) at these sites remains challenging. In this study, LNPsD18, a cationic liposomal formulation that targets and enhances APC uptake at both sites without modifying the targeting ligands is developed. By co-delivering tumor-specific neoantigens and a cholesterol-coupled toll-like receptor 9 (TLR9) agonist within LNP-vaxD18, an approximately 60-fold increase in dendritic cell uptake compared to neoantigen-adjuvant mixtures is achieved. Intravenous administration of the liposome-neoantigen peptide vaccine targets both the spleen and the tumor, boosting splenic DC activation, increasing M1-type tumor-associated macrophages, and elevating tumor cytokine levels. This reshapes the tumor microenvironment, enhancing IFN-γ-producing CD8+ T cells and TCF1+CD8+ T cells within tumors. These outcomes significantly inhibit established tumor growth compared to nontargeted lipid-based nanovaccine formulations, resulting in improved survival in orthotopic hepatocellular carcinoma and colorectal cancer models. The findings highlight the importance of targeting APCs in both the spleen and tumors to optimize the therapeutic efficacy of liposome-neoantigen vaccines in cancer treatment.
Collapse
Affiliation(s)
- Yu Xu
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510060P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhou510060P. R. China
| | - Bing Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Fujian Agriculture and Forestry UniversityFuzhou350002P. R. China
| | - Yue Huang
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510060P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhou510060P. R. China
| | - JianPing Liao
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510060P. R. China
| | - Chenyi Wu
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510060P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhou510060P. R. China
| | - Chenxi Zhou
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Zishi Kang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Shiyang Jiang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Bing‐Chen Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Da Zhang
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510060P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhou510060P. R. China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Ruihua Xu
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhou510060P. R. China
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen UniversityGuangzhou510060P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- CAS Key Laboratory of Design and Assembly of Functional NanostructuresFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhou350002P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Feng Wang
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen UniversityGuangzhou510060P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal CancerChinese Academy of Medical SciencesGuangzhou510060P. R. China
| |
Collapse
|
234
|
Sohail SK. Natural Products as Modulators of miRNA in Hepatocellular Carcinoma: A Therapeutic Perspective. J Gene Med 2025; 27:e70019. [PMID: 40296860 DOI: 10.1002/jgm.70019] [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: 03/17/2025] [Revised: 04/07/2025] [Accepted: 04/20/2025] [Indexed: 04/30/2025] Open
Abstract
Hepatocellular carcinoma (HCC) continues to pose a substantial worldwide health concern, marked by elevated mortality rates and restricted therapeutic alternatives. Recent studies have highlighted the potential of natural compounds as therapeutic agents in cancer management. This review focuses on the diagnostic and prognostic potential of microRNAs (miRNAs) as biomarkers in HCC, alongside the therapeutic promise of natural products. We explore the intricate role of miRNAs in the pathogenesis of HCC, detailing their regulatory functions in cellular processes such as proliferation, apoptosis, and metastasis. Additionally, we discuss the emerging evidence supporting the use of natural compounds, including phytochemicals, in modulating miRNA expression and their potential synergistic effects with conventional therapies. Key miRNAs discussed include miR-21, an oncogenic factor that promotes tumor growth by targeting the tumor suppressor phosphatase and tensin homolog (PTEN); miR-34a, which enhances apoptosis and may improve treatment efficacy when combined with c-MET inhibitors; miR-203, whose downregulation correlates with poor outcomes and may serve as a prognostic marker; miR-16, which acts as a tumor suppressor and has diagnostic potential when measured alongside traditional markers like alpha-fetoprotein (AFP); and miR-483-3p, associated with resistance to apoptosis and tumor progression. By integrating insights from recent studies, this review aims to highlight the dual role of miRNAs as both biomarkers and therapeutic targets, paving the way for enhanced diagnostic strategies and novel treatment modalities in HCC management.
Collapse
|
235
|
Osman H, Wang F, Zou G, Zhang D, Bai X, Jiang T, Wang Y. Antibacterial and osteogenic gain strategy on titanium surfaces for preventing implant-related infections. Colloids Surf B Biointerfaces 2025; 249:114489. [PMID: 39787742 DOI: 10.1016/j.colsurfb.2024.114489] [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: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/12/2025]
Abstract
Infection and insufficient osseointegration are the primary factors leading to the failure of titanium-based implants. Surface coating modifications that combine both antibacterial and osteogenic properties are commonly employed strategies. However, the challenge of achieving rapid antibacterial action and consistent osteogenesis with these coatings remains unresolved. In this study, a functional composite coating (PDA/PPy@Cu/Dex) was prepared on titanium surfaces using layer-by-layer self-assembly and electrochemical deposition techniques. The hydroxyl groups grafted by polydopamine's (PDA) self-polymerization and the enhanced conductivity and uniform electric field distribution provided by polypyrrole (PPy) allowed for the even dispersion of copper nanoparticles and dexamethasone (Dex) on the titanium surface. This synergistically coupled the photothermal ion antibacterial properties of copper nanoparticles with the osteogenic promotion of dexamethasone. In vitro antibacterial experiments revealed that the heat generated by photothermal effects and reactive oxygen species enhanced the antibacterial activity of copper ions, reducing the antibacterial time to six h and achieving antibacterial enhancement. In vitro cell experiments showed that the long-term slow release of copper ions and dexamethasone enhanced the osteogenic differentiation of stem cells, thereby achieving osteogenic benefits. Moreover, in vivo toxicity experiments demonstrated that the composite coating had no adverse effects on normal tissues. Therefore, the antibacterial and osteogenic enhancement strategy for titanium surfaces presented in this study offers a new potential approach for preventing implant-associated infections.
Collapse
Affiliation(s)
- Henigul Osman
- College of Chemical Engineering, Xinjiang Normal University, 102 Xinyi Road, Urumqi 830054, P.R. China
| | - Fan Wang
- Department of Orthopaedics, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P.R. China
| | - Guoming Zou
- College of Chemical Engineering, Xinjiang Normal University, 102 Xinyi Road, Urumqi 830054, P.R. China
| | - Dong Zhang
- College of Chemical Engineering, Xinjiang Normal University, 102 Xinyi Road, Urumqi 830054, P.R. China
| | - Xi Bai
- College of Chemical Engineering, Xinjiang Normal University, 102 Xinyi Road, Urumqi 830054, P.R. China
| | - Tao Jiang
- Laboratory Animal Center of Xinjiang Medcial University, 393 Xinyi Road, Urumqi 830011, P.R. China.
| | - Yingbo Wang
- College of Chemical Engineering, Xinjiang Normal University, 102 Xinyi Road, Urumqi 830054, P.R. China.
| |
Collapse
|
236
|
Miri H, Rahimzadeh P, Hashemi M, Nabavi N, Aref AR, Daneshi S, Razzaghi A, Abedi M, Tahmasebi S, Farahani N, Taheriazam A. Harnessing immunotherapy for hepatocellular carcinoma: Principles and emerging promises. Pathol Res Pract 2025; 269:155928. [PMID: 40184729 DOI: 10.1016/j.prp.2025.155928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
HCC is considered as one of the leadin causes of death worldwide, with the ability of resistance towards therapeutics. Immunotherapy, particularly ICIs, have provided siginficant insights towards harnessing the immune system. The present review introduces the concepts and possibilities of immunotherapy for HCC treatment, emphasizing its underlying mechanisms and capacity to enhance patient results, focusing on both pre-clinical and clinical insights. The functions of TME and immune evasion mechanisms typical of HCC would be evaluated along with how contemporary immunotherapeutic approaches are designed to address these challenges. Furthermore, the clinical application of immunotherapy in HCC is discussed, emphasizing recent trial findings demonstrating the effectiveness and safety of drugs. In addition, the problems caused by immune evasion and resistance would be discussed to increase potential of immunotherapy along with combination therapy.
Collapse
Affiliation(s)
- Hossein Miri
- Faculty of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX, Inc, Boston, MA, USA
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University Of Medical Sciences, Jiroft, Iran
| | - Alireza Razzaghi
- Social Determinants of Health Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Maryam Abedi
- Department of Pathology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Safa Tahmasebi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
237
|
Lang J, Liu Q, Ji R, Qiu M, Wang S, Liu Q, Li D, Chen P, Xiong Z. Complete remission of a high-risk, locally advanced cervical cancer with para-aortic lymph node metastases treated with first-line tislelizumab plus bevacizumab combined with chemotherapy followed by radiotherapy with maintenance therapy: a case report. Front Immunol 2025; 16:1573202. [PMID: 40375998 PMCID: PMC12078261 DOI: 10.3389/fimmu.2025.1573202] [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: 02/08/2025] [Accepted: 04/08/2025] [Indexed: 05/18/2025] Open
Abstract
Newly diagnosed cervical cancer with metastatic para-aortic lymph node (PALN) involvement is associated with a significantly poor prognosis, with distant metastasis being the predominant pattern of treatment failure. The programmed cell death receptor-1 (PD-1) pathway has garnered considerable attention due to its role in enabling tumor cells to evade immune surveillance by eliciting the immune checkpoint response of T cells, rendering them highly refractory to conventional chemotherapy. The National Comprehensive Cancer Network (NCCN) guidelines currently recommend pembrolizumab for locally advanced cervical cancer patients positive for PD-L1 (CPS ≥1), as determined by an FDA-approved assay. Tislelizumab, an anti-PD-1 monoclonal IgG4 antibody, has been investigated in hematological malignancies and advanced solid tumors. Nevertheless, literature on regimens incorporating tislelizumab for the treatment of locally advanced cervical cancer is scarce. Herein, we present a case of a newly diagnosed high-risk, locally advanced cervical cancer patient with PALN metastases and low PD-L1 expression, treated with a combination of tislelizumab, bevacizumab, and a platinum-containing chemotherapy regimen followed by radiotherapy with maintenance therapy, resulting in a notable extension of progression-free survival.
Collapse
Affiliation(s)
- Juan Lang
- Department of Pathology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Qianqian Liu
- Department of Radiation Oncology, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Rong Ji
- Department of Radiation Oncology, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Miao Qiu
- Department of Radiation Oncology, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Siben Wang
- Department of Radiation Oncology, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Qingmeng Liu
- Department of Pathology, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Dapeng Li
- Department of Radiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ping Chen
- Department of Obstetrics and Gynecology, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Zhongkui Xiong
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
- Department of Radiation Oncology, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
238
|
Chen X, Zhang S, Wang C, Chao T, Ren J, Gao F, Liu Z, Peng R. Mineralized Bacteria as a Potent Vaccine Against Staphylococcus aureus Infections. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412279. [PMID: 40178324 DOI: 10.1002/smll.202412279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/07/2025] [Indexed: 04/05/2025]
Abstract
Staphylococcus aureus (S. aureus) as common Gram-positive pathogenic bacteria, causes local and systemic infections, including sepsis and bacteremia. In particular, the high prevalence of drug-resistant S. aureus further complicates the post-infection treatment. Highly effective S. aureus vaccines are urgently desired. Herein, a novel S. aureus vaccine (MnO2@FS) is developed via biomineralizing manganese dioxide (MnO2) on formaldehyde-fixed S. aureus (FS). In such vaccine, with FS to induce bacteria-specific immune responses, MnO2 via releasing Mn2+ can activate the cyclic GMP-AMP synthase-stimulator of interferon gene (cGAS-STING) pathway and innate immunity, which would be rather helpful to enhance immune responses against bacterial infections. It is found that bone marrow-derived dendritic cells (BMDCs) treated with MnO2@FS show higher FS and manganese uptake, and enhanced cytokine secretions. In mice, after being immunized with MnO2@FS, the level of S. aureus-specific antibody is significantly improved compared with FS and simple mixture of FS and MnO2 (FS+MnO2). Furthermore, MnO2@FS immunized mice can clear infected bacteria faster and showing higher survival rate in lethal models, outperforming FS and FS+MnO2 immunizations. In addition, the vaccine effectively controls abscess development in a hospital-acquired S. aureus infection model. This study thus presents a new strategy for the construction of highly potent yet safe bacterial vaccines.
Collapse
Affiliation(s)
- Xiaojing Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Shiyuan Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Chenya Wang
- InnoBM Pharmaceuticals Co, Itd., Suzhou, Jiangsu, 215123, China
| | - Ting Chao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jiacheng Ren
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Feng Gao
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, 215009, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Rui Peng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
239
|
Zhang J, Yu Q, Zhu W, Sun X. Recent advances in the role of circRNA in cisplatin resistance in tumors. Cancer Gene Ther 2025; 32:497-506. [PMID: 40148680 DOI: 10.1038/s41417-025-00899-4] [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: 12/27/2024] [Revised: 03/08/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Cancer remains a major threat to human health, with chemotherapy serving as one of the main treatment strategies to alleviate patient suffering. However, prolonged chemotherapy often leads to the development of drug resistance, complicating treatment outcomes. Cisplatin, a commonly utilized chemotherapeutic agent, demonstrates efficacy against a range of cancers but frequently encounters resistance, posing a significant challenge in tumor management and prognosis. Drug resistance not only facilitates tumor progression but also reduces survival rates, highlighting the urgent need for innovative strategies to overcome this issue. In recent years, non-coding RNAs, particularly circular RNAs (circRNAs), have gained attention in cancer therapy due to their stability and specificity. Moreover, an increasing number of studies have reported that circRNAs are involved in cisplatin resistance across various types of cancer. This paper primarily reviews the mechanisms and roles of circRNA in mediating cisplatin resistance over the past 3 years. These findings highlight circRNAs as promising therapeutic targets for overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Jiawen Zhang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qiwen Yu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Weijin Zhu
- Department of Clinical Laboratory Medicine, Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu, China
| | - Xiaochun Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
240
|
Yong GY, Kamkaew A, Kue CS. Synergistic approach of PEGylated photothermal agent and immunomodulator in cancer immunotherapy. Nanomedicine (Lond) 2025; 20:967-983. [PMID: 40214079 PMCID: PMC12051527 DOI: 10.1080/17435889.2025.2489342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 04/02/2025] [Indexed: 05/02/2025] Open
Abstract
Photothermal therapy (PTT) utilizes photothermal agents (PTAs) to generate heat at the local tumor site that leads to ablation upon photoirradiation at a specific wavelength of light. Currently, most of the available PTAs have weak tumor selectivity and depositing ability, which leads to poor therapeutic outcomes. PEGylation of PTAs improves therapeutic outcomes, prolongs systemic circulation time, enhances tumor accumulation, and reduces the risk of clearance by the immune system. This paper reviews the recent developments of PEGylated PTAs in photothermal cancer therapy from 2019 to 2023, highlighting their antitumour efficacy and immune response post-therapy with immune agents, current challenges and strategies. This review aims to foster knowledge dissemination on the application of nanomedicine in photothermal cancer therapy from an immunological perspective and to encourage the clinical translation of these nanomaterials.
Collapse
Affiliation(s)
- Gong Yi Yong
- School of Graduate Studies, Management and Science University, Shah Alam, Malaysia
| | - Anyanee Kamkaew
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Chin Siang Kue
- Faculty of Health and Life Sciences, Management and Science University, Shah Alam, Malaysia
| |
Collapse
|
241
|
Bai L, Liu X, Yuan Z, Xu G, Li X, Wan Z, Zhu M, Liang X, Li P, Lan Q, Yu H, Tang G, Huang M, Peng S, Lin J, Wang X, Luo Y, Wei G. Activation of IL-2/IL-2R pathway by Hedyotis diffusa polysaccharide improves immunotherapy in colorectal cancer. Int J Biol Macromol 2025; 306:141013. [PMID: 39954887 DOI: 10.1016/j.ijbiomac.2025.141013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Colorectal cancer (CRC) is a prevalent and highly malignant tumor with a limited response to immune checkpoint inhibitor-based immunotherapy. There is an urgent need for novel immunomodulatory agents to enhance the immunotherapeutic response in CRC. Hedyotis diffusa, known for its immunomodulatory properties, has long been utilized as an adjunct in cancer treatment, positioning it as a potential source for discovering new tumor immunomodulators. In this study, we identified a polysaccharide derived from Hedyotis diffusa (HDP), comprising six monosaccharides: rhamnose, arabinose, galactose, glucose, xylose, and mannose. When combined with PD-1 and CTLA-4 inhibitors, HDP can boost systemic immunity in mice to enhance the effectiveness of immune checkpoint inhibitors in CRC therapy. HDP significantly increases the infiltration of CD4+ and CD8+ T cells into tumor microenvironment and upregulates the expression of key effector molecules derived from cytotoxic T cells. Mechanistic studies reveal that HDP activates the IL-2/IL-2R axis by upregulating IL-2 production and the expression of IL-2 receptor subunits, thereby promoting T cell proliferation. Collectively, this research introduces an innovative strategy to improve the efficacy of tumor immunotherapy by harnessing the immunomodulatory potential of polysaccharides. It also directs a roadmap for developing HDP as a promising immunomodulator for CRC treatment.
Collapse
Affiliation(s)
- Liangliang Bai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Xiaoxia Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Ze Yuan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Gaopo Xu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xuan Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Zhongxian Wan
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei 445000, China
| | - Mingxuan Zhu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xiaoxia Liang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Peisi Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Qiqian Lan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Huichuan Yu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Guannan Tang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Mingzhe Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Shaoyong Peng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Jinxing Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xiaolin Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Yanxin Luo
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Gang Wei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
242
|
Mossburg KJ, Barragan D, O NH, Kian AC, Maidment ADA, Cormode DP. Emerging nanoparticle-based x-ray imaging contrast agents for breast cancer screening. Nanomedicine (Lond) 2025; 20:1149-1166. [PMID: 40261216 PMCID: PMC12068350 DOI: 10.1080/17435889.2025.2496129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025] Open
Abstract
Breast cancer is one of the most common types of cancer, however, preventive screening has contributed to a significant reduction in mortality over the past four decades. The first-line screening methods for breast cancer, such as mammography and tomosynthesis, are x-ray-based modalities. Unfortunately, their cancer detection rates are low in patients with dense breasts. These, and other high-risk women, are now encouraged to receive supplemental screening. The supplemental imaging methods are diverse, including ultrasound, MRI, nuclear imaging, and X-ray-based modalities such as breast CT and contrast-enhanced mammography/tomosynthesis. Due to their low cost and wide availability, x-ray-based modalities see significant clinical use worldwide. These techniques benefit from the use of contrast agents, which are currently iodinated small molecules designed for other purposes. Consequently, developing new contrast agents that are specifically for breast cancer screening is of interest. This review describes these modalities and the nanoparticle-based contrast agents being researched for their enhanced performance. The relevant parameters for nanoparticle-based contrast agent design are evaluated, including contrast generation and potential biointeractions. Iodinated agents are discussed for comparison. Nanoparticles covered include silver sulfide, silver telluride, gold, and bismuth sulfide-based agents, among others. Finally, perspectives on future developments in this field are offered.
Collapse
Affiliation(s)
- Katherine J. Mossburg
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Diego Barragan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathaniel H. O
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pharmaceutical Sciences, St. Joseph’s University, Philadelphia, PA, USA
- Department of Physics, St. Joseph’s University, Philadelphia, PA, USA
| | - Andrea C. Kian
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew D. A. Maidment
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - David P. Cormode
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
243
|
Kong S, Li J, Pan X, Zhao C, Li Y. ZNF384 and m6A methylation promote the progression of hepatocellular carcinoma by regulating the interaction between LINC00342 and DAPK1. Cell Signal 2025; 129:111666. [PMID: 39961407 DOI: 10.1016/j.cellsig.2025.111666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/05/2024] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with high morbidity and mortality. Many lncRNAs play important regulatory roles in the pathogenesis of HCC, but the mechanism of action of LINC00342 in the progression of HCC remains unclear. In this study, we assessed 24 pairs of HCC tissues and adjacent normal tissues as well as HCC cells and a nude mouse model of HCC. Gene and protein expression was evaluated by flow cytometry, CCK-8, RIP, colony formation assay, and TUNEL staining. This study revealed that LINC00342 was highly expressed in HCC tissues and cells. LINC00342 knockdown significantly inhibited the proliferation and migration of HCC cells, promoted apoptosis, inhibited tumor growth in vivo, and increased the sensitivity of HCC cells to cisplatin. The opposite effect was observed in LINC00342-overexpressing cells. Mechanistically, ZNF384 and m6A methylation can promote the transcription and stability of LINC00342, and LINC00342 can bind to DAPK1, which inhibits Cyt C release and the activation of caspase family proteins to accelerate HCC progression. Our study indicated that the inhibition of LINC00342 expression may represent a new breakthrough for HCC treatment.
Collapse
Affiliation(s)
- Shujia Kong
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Jiaxun Li
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Xin Pan
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Chen Zhao
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Yanwen Li
- Intensive Care Unit, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China.
| |
Collapse
|
244
|
Qiu SQ, He XF, Liang XL, Shi GY, Zhao ML, Li F, Wu ZY, Tian J, Zhai TT, Du Y. GLUT1 as a generic biomarker enables near-infrared fluorescence molecular imaging guided precise intraoperative tumor detection in breast cancer. Eur J Nucl Med Mol Imaging 2025; 52:2171-2186. [PMID: 39833507 DOI: 10.1007/s00259-025-07095-4] [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: 10/08/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE Precise tumor excision is important but challenging in breast-conserving surgery (BCS). Tumor-specific fluorescence imaging may be used for intraoperative tumor detection and, therefore, to guide precise tumor excision. The aims of this study are to develop a glucose transporter 1 (GLUT1)-targeted near-infrared fluorescence tracer and evaluate its accuracy for breast cancer detection using fresh surgical breast specimens. METHODS Bioinformatic analysis was performed to compare GLUT1 expression between breast cancer and normal breast tissues. A GLUT1-targeted fluorescence imaging tracer WZB117-CY7.5 was developed. In combination with fluorescence imaging (FMI), its binding specificity to GLUT1 was examined in in vitro breast cancer cell experiments, in vivo 4T1 breast tumor-bearing mouse models, and 60 freshly resected human breast tumor tissues. The diagnostic accuracy of WZB117-CY7.5, was evaluated in fresh specimens derived from 60 patients diagnosed with breast cancer. RESULTS GLUT1 expression is higher in breast cancer tissues compared with normal tissues. WZB117-CY7.5 specifically bound to breast cancer cells in in vitro cell experiments and accumulated in tumor areas in a 4T1 tumor-bearing mice after intravenous injection by FMI. Moreover, WZB117-CY7.5 specifically bound to freshly resected human breast cancer and demonstrated excellent diagnostic performance in discriminating breast cancer, irrespective of cancer subtype, from normal breast tissue on fresh surgically resected breast tissues. CONCLUSIONS WZB117-CY7.5 showed high accuracy in intraoperative breast cancer detection, irrespective of the cancer subtype. This highlights its potential for clinical applications as a generic tracer for fluorescence image-guided surgery (FIGS) in BCS and fluorescence image-guided pathology for tissue sampling.
Collapse
Affiliation(s)
- Si-Qi Qiu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, No.114 Waima Road, Shantou, 515041, Shantou, China
- Clinical Research Center, Shantou Central Hospital, Shantou, 515041, China
| | - Xiao-Feng He
- Shantou University Medical College, Shantou, 515041, China
| | - Xiao-Long Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Guang-Yuan Shi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No.95 Zhongguancun East Road, Hai Dian District, Beijing, 100190, China
| | - Meng-Long Zhao
- Clinical Research Center, Shantou Central Hospital, Shantou, 515041, China
| | - Fan Li
- Biobank, Shantou Central Hospital, Shantou, 515041, China
| | - Zhi-Yong Wu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, No.114 Waima Road, Shantou, 515041, Shantou, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No.95 Zhongguancun East Road, Hai Dian District, Beijing, 100190, China.
- Beijing Advanced Innovation Centre for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, 100191, China.
| | - Tian-Tian Zhai
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, No.7 Raoping Road, Jinping District, Shantou, 515041, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No.95 Zhongguancun East Road, Hai Dian District, Beijing, 100190, China.
- The University of Chinese Academy of Sciences, Beijing, 100080, China.
| |
Collapse
|
245
|
Tan J, Ding B, Chen H, Meng Q, Li J, Zhang W, Yang Z, Ma X, Han D, Yang M, Zheng P, Ma P, Lin J. Gallium-Magnesium Layered Double Hydroxide for Elevated Tumor Immunotherapy Through Multi-Network Synergistic Regulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2501256. [PMID: 40190140 DOI: 10.1002/adma.202501256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/12/2025] [Indexed: 05/28/2025]
Abstract
Immunotherapeutic efficacy is often limited by poor immunogenicity, immunosuppressive tumor microenvironment (TME), and cytoprotective mechanisms, leading to low immune activation. To this end, here, L-amino acid oxidase (LAAO) loaded gallium-magnesium layered double hydroxide (MG-LAAO) is prepared for significantly enhanced tumor immunotherapy through multi-network synergistic regulation. First, MG-LAAO induces tumor cell pyroptosis by initiating caspase-1/GSDMD and caspase-3/GSDME pathways, further triggering immunogenic cell death (ICD). Then the released Ga3+ induces mitochondrial iron overload, resulting in ferroptosis. In addition, MG-LAAO also hinders autophagy of tumor cells, and reshapes the immunosuppressive tumor microenvironment (TME) by neutralizing H+ and inhibiting lactic acid accumulation, thus destroying the cytoprotective mechanism and avoiding immune escape. Furthermore, this multi-network synergy further activates the cGAS-STING signaling pathway, generating powerful antitumor immunotherapy. This work highlights the critical role of synergies between autophagy block, pyroptosis, ferroptosis, and ICD in tumor immunotherapy, demonstrating the important role of this multi-network synergy in effectively overcoming immunosuppressive TME and enhancing immunogenicity. In particular, the mechanism of gallium-induced pyroptosis is revealed for the first time, providing theoretical support for the design of new materials for tumor immunotherapy in the future.
Collapse
Affiliation(s)
- Jia Tan
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Hao Chen
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Jing Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Wenying Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Zhuang Yang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Xinyu Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Di Han
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Mingkai Yang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Pan Zheng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- Key Laboratory of Superlight Materials and Surface Technology of Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
246
|
Tang Y, Yuan F, Cao M, Ren Y, Li Y, Yang G, Zhong Z, Liang H, Xiong Z, He Z, Lin N, Deng M, Yao Z. CircRNA-mTOR Promotes Hepatocellular Carcinoma Progression and Lenvatinib Resistance Through the PSIP1/c-Myc Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410591. [PMID: 40231634 PMCID: PMC12120768 DOI: 10.1002/advs.202410591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/26/2024] [Indexed: 04/16/2025]
Abstract
Circular RNAs (circRNAs) are crucial regulators of targeted drug resistance in hepatocellular carcinoma (HCC). However, the specific mechanisms underlying resistance that significantly hampers the effectiveness of HCC treatments remain unclear. Here, it is found that circRNA-mTOR is highly expressed in HCC and strongly correlated with patient prognosis. Furthermore, circRNA-mTOR enhances the stemness of HCC cells, thereby promoting the progression of HCC and contributing to lenvatinib resistance. Mechanistically, circRNA-mTOR promotes the nuclear translocation of the RNA-binding protein (RBP) PC4 and SRSF1 interacting protein 1 (PSIP1) through specific binding. The enhancement of HCC cell stemness by circRNA-mTOR occurs via the PSIP1/c-Myc signaling pathway, ultimately driving HCC progression and lenvatinib resistance. This study highlights the important role of circRNA-mTOR in HCC progression and the maintenance of lenvatinib resistance and underscores its potential as a biomarker for the diagnosis and prognosis of HCC. In conclusion, this study provides an experimental foundation for targeted drug therapy in HCC and offers novel insights, perspectives, and methodologies for understanding the development and occurrence of this disease. These findings are significant for the development of new diagnostic and therapeutic markers for HCC, with the ultimate goal of reducing drug resistance.
Collapse
Affiliation(s)
- Yongchang Tang
- Department of Hepatobiliary SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
- Department of General SurgeryQilu HospitalShandong UniversityJinan250012China
| | - Feng Yuan
- Department of Hepatobiliary SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
- Department of Hepatobiliary SurgeryThe First Affiliated HospitalGuangzhou Medical UniversityGuangzhou510120China
| | - Mingbo Cao
- Department of Hepatobiliary SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Yupeng Ren
- Department of Hepatobiliary SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Yuxuan Li
- Department of Hepatobiliary SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Gaoyuan Yang
- Department of Hepatobiliary SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Zhaozhong Zhong
- Department of Hepatobiliary SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
- Department of Kidney TransplantationThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Hao Liang
- Department of Hepatobiliary and Pancreatic SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Zhiyong Xiong
- Department of Hepatobiliary and Pancreatic SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Zhiwei He
- Department of Hepatobiliary SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Nan Lin
- Department of Hepatobiliary SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Meihai Deng
- Department of Hepatobiliary SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Zhicheng Yao
- Department of Hepatobiliary and Pancreatic SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| |
Collapse
|
247
|
Qian J, Li X, Ruan H, Du Z, Wei S, Sun Y. Design and development of drug delivery nanocarriers based on liquid-liquid phase separation, improved stability, cell-penetration and anti-cancer effect. Int J Biol Macromol 2025; 307:142023. [PMID: 40086555 DOI: 10.1016/j.ijbiomac.2025.142023] [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: 01/02/2025] [Revised: 03/02/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Liquid-liquid phase separation (LLPS) of nuclear pore complex (NPC) with nuclear transport proteins (NTPs) via intrinsically disordered regions (IDRs) plays a crucial role in the nucleocytoplasmic transport. The development of efficient targeted delivery systems based on LLPS has attracted widespread attention. Here, we developed nanocarriers of casein peptides, a natural intrinsically disordered proteins (IDPs), modified with fatty acids of different alkyl chains (C10-C18) and decorated by shellac for highly effective drug delivery and cancer therapy. The curcumin (Cur)-loading nanocarriers (CSLNCs) showed excellent stability and dispersity in the natural environment over 30 days, with Cur encapsulation efficiency and loading capacity of ~90 % and ~57 %. Electron microscope (EM) indicated an aggregated homogeneous elliptical shape of CSLNCs(C10) and the morphology of CSLNCs(C18) transited to a distributed cubic shape. CSLNCs(C10, C12, C14 and C18) exhibited cytotoxicity against human lung adenocarcinoma NCI-H1975 cells with an IC50 value of 17.5 μM, 17.3 μM, 10.2 μM and 19.3 μM after 24 h of incubation, respectively. CSLNCs were also found to inhibit the cell wound healing with a migration rate of 12.72 %, 10.93 %, 4.28 % and 13.62 %, respectively. CSLNCs especially increased the percentage of late apoptotic cells. As indications of confocal microscopy, the fluorescence intensities of NCI-H1975 cells were enhanced with a cytosolic distribution and noticeably florescence in the nucleus after 0.5 h of incubation CSLNCs. CSLNCs treated cells adopted a rounded morphology with a dramatic reduction in fluorescence intensity after 1 h of incubation. Among CSLNCs, CSLNCs(C14) improved considerably the cytotoxicity activity and intercellular localization in the nucleus. The cell-penetration ability was also confirmed by the binding of CSLNCs in a model bicelles membrane system composed of DMPC and DHPC investigated by 1H NMR. It was proposed that CSLNCs with cell-penetrating and nuclear targeting performance may regulate the LLPS of nuclear pore complex and thus improved its nuclear penetration and cytotoxic activity.
Collapse
Affiliation(s)
- Jingya Qian
- College of Vocational and Technical Education, Yunnan Normal University, Kunming, Yunnan, China
| | - Xiujuan Li
- Pharmaceutical Department, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Hefei Ruan
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhongyao Du
- Yunnan Key Laboratory of Modern Separation Analysis and Substance Transformation, College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, Yunnan, China
| | - Song Wei
- Tumor Precise Intervention and Translational Medicine Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China.
| | - Yang Sun
- Yunnan Key Laboratory of Modern Separation Analysis and Substance Transformation, College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, Yunnan, China.
| |
Collapse
|
248
|
Ramirez AB, Lazenby RA. Fabricating Reproducible, Reversible, and High Signal Change Aptasensors with Gold-Modified Nanopipettes. ACS APPLIED MATERIALS & INTERFACES 2025; 17:24877-24886. [PMID: 40257359 DOI: 10.1021/acsami.4c22935] [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: 04/22/2025]
Abstract
Aptamer-functionalized nanopipettes are an emerging class of biosensors for the label-free detection of specific molecules. While various strategies exist for immobilizing single-stranded DNA aptamers onto the inner walls of glass nanopipettes, the impact of the fabrication method on sensor sensitivity, signal change, reproducibility, and reliability remains unexplored. In this study, we compared three fabrication methods and found that sensors fabricated using gold nanoparticles (AuNPs) synthesized within the nanopipettes produced the most reproducible results while also allowing control over the modification process. In contrast, two other aptamer immobilization methods, which relied on multistep polymer coatings with aminated or thiolated aptamer coupling, were hindered by water sensitivity and uneven polymer deposition, resulting in inconsistent sensor responses. Using the AuNP-coated nanopipettes, we successfully fabricated numerous sensors of varying sizes, demonstrating that smaller nanopipettes produce greater signal changes. Sensors constructed using glass nanopipettes with diameters ranging from 22 to 30 nm exhibited large signal changes (>40%) when AuNP synthesis produced particles near the tip opening without causing blockage. However, we also observed sensors with signal changes that were significantly lower (using the same-sized glass nanopipettes), which we attributed to either minimal Au present at the tip or conversely when Au significantly blocked the probe. These results highlight the critical role of fabrication methods in maximizing the signal change, enhancing the reproducibility, and identifying how and why sensors fail. This work aims to facilitate the broader adoption of aptamer-functionalized nanopipettes in analytical sensing applications.
Collapse
Affiliation(s)
- Ana B Ramirez
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Robert A Lazenby
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| |
Collapse
|
249
|
Li L, Li M, Qiu Y, Wang S, Dong Y. Aptamers capable of simultaneously identifying multiple targets and corresponding applications in medical diagnosis-A review. Int J Biol Macromol 2025; 311:143666. [PMID: 40316072 DOI: 10.1016/j.ijbiomac.2025.143666] [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: 01/10/2025] [Revised: 04/20/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025]
Abstract
Aptamers, a unique class of nucleic acid sequences recognized for their specific binding capabilities, have found widespread application in biomedical field. While traditional aptamers are typically designed to target a single molecule recognition, recent attention has been directed towards multifunctional aptamers capable of simultaneously identifying multiple targets. In this review, the latest advancements in multifunctional aptamers and their applications in medical diagnosis are presented for the first time. This review focuses on the following essential aspects, including methods employed for developing multifunctional aptamers, detailed characteristics of these aptamers, practical applications across diverse diagnostic scenarios, and in-depth discussions on critical aspects of their design and utility. To conclude, future perspectives are provided to drive further development and broader application of multifunctional aptamers in the biomedical domain.
Collapse
Affiliation(s)
- Ling Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Menglei Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Yinghua Qiu
- Center for Molecular Diagnostics and Precision Medicine, Institute for Molecular Medicine and Infectious Disease, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia 19102, USA
| | - Sai Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, PR China
| | - Yiyang Dong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| |
Collapse
|
250
|
Yu X, Zhang Q, Wang L, Zhang Y, Zhu L. Engineered nanoparticles for imaging and targeted drug delivery in hepatocellular carcinoma. Exp Hematol Oncol 2025; 14:62. [PMID: 40307921 PMCID: PMC12044934 DOI: 10.1186/s40164-025-00658-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/18/2025] [Indexed: 05/02/2025] Open
Abstract
Liver cancer, notably hepatocellular carcinoma (HCC), poses a significant global health burden due to its high fatality rates. Conventional antitumor medications face challenges, including poor targeting, high toxicity, and drug resistance, leading to suboptimal clinical outcomes. This review focused on nanoparticle use in diagnosing and delivering medication for HCC, aiming to advance the development of nanomedicines for improved treatment outcomes. As an emerging frontier science and technology, nanotechnology has shown great potential, especially in precision medicine and personalized treatment. The success of nanosystems is attributable to their smaller size, biocompatibility, selective tumor accumulation, and lower toxicity. Nanoparticles, as a central part of nanotechnology innovation, have emerged in the field of medical diagnostics and therapeutics to overcome the various limitations of conventional chemotherapy, thus offering promising applications for improved selectivity, earlier and more precise diagnosis of cancers, personalized treatment, and overcoming drug resistance. Nanoparticles play a crucial role in drug delivery and imaging of HCC, with the body acting as a delivery system to target and deliver drugs or diagnostic reagents to specific organs or tissues, helping to accurately diagnose and target therapies while minimizing damage to healthy tissues. They protect drugs from early degradation and increase their biological half-life.
Collapse
Affiliation(s)
- Xianzhe Yu
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, No. 10 Qinyun Nan Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Qin Zhang
- Department of Postgraduate Students, West China School of Medicine/West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Leibo Wang
- Department of Surgery, Beijing Jishuitan Hospital Guizhou Hospital Guiyang, Guiyang, 550000, Guizhou, The People's Republic of China
| | - Yan Zhang
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Lingling Zhu
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|