301
|
Molino J, Ibrahim M, Serra R, de Tristán S. Optimization of seebeck coefficients in polyaniline-doped manganese dioxide nanocomposites. PLoS One 2025; 20:e0321385. [PMID: 40238809 PMCID: PMC12002486 DOI: 10.1371/journal.pone.0321385] [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: 08/02/2024] [Accepted: 03/04/2025] [Indexed: 04/18/2025] Open
Abstract
Polyaniline (PANI) and PANI-MnO2 composites were synthesized via a chemical route with varying manganese dioxide (MnO2) content, specifically 5wt% and 15wt%. X-ray diffraction (XRD) confirmed the structural formation of both PANI and PANI-MnO2 composites. The direct current conductivity was measured, showing an increase with temperature: at 393K, pure PANI had a conductivity of 2.25 × 10-4 S/cm, which increased significantly in the composites, reaching 9.03 × 10-4 S/cm for the 15wt% MnO2 composite. The Seebeck coefficient also increased with temperature and MnO2 concentration, achieving a maximum value of 52 mV K-1 at 373K for the 15wt% MnO2 composite. These results indicate that the synthesized PANI- MnO2 composites exhibit semiconducting behavior with improved thermoelectric properties, making them promising candidates for applications in thermoelectric devices such as generators and thermopiles. The study highlights the potential of these materials in enhancing the efficiency of thermoelectric energy conversion.
Collapse
Affiliation(s)
- Jay Molino
- Universidad Especializada de las Américas (UDELAS), Faculty of Biosciences and Public Health, Biomedical Engineering, Centro I+D+i de Biotecnología, Energías Verdes y Cambio Climático, Albrook, Paseo de La Iguana, Republic of Panama
- Sistema Nacional de Investigación (SNI), SENACYT, Panama City, Republic of Panama
| | - Muhammad Ibrahim
- Faculty of Engineering and Science, Bahauddin Zakariya University – BZU, Punjab, Pakistan
| | - Rolando Serra
- Departamento de Física, Universidad Tecnológica de La Habana José Antonio Echeverría, La Habana, Cuba
| | - Svetlana de Tristán
- Universidad Especializada de las Américas (UDELAS), Faculty of Biosciences and Public Health, Biomedical Engineering, Centro I+D+i de Biotecnología, Energías Verdes y Cambio Climático, Albrook, Paseo de La Iguana, Republic of Panama
| |
Collapse
|
302
|
Mahata R, Manna S, Modak M, Choudhury SM. A review on the advancement of polydopamine (PDA)-based nanomaterials for cancer treatment. Med Oncol 2025; 42:165. [PMID: 40237855 DOI: 10.1007/s12032-025-02678-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/11/2025] [Indexed: 04/18/2025]
Abstract
The significance of cancer treatment research lies in addressing the high incidence of cancer, overcoming treatment challenges, and mitigating the harsh side effects of chemotherapeutic agents. Currently, nanotechnology is garnering significant attention for its potential applications in diagnostics and drug delivery, offering innovative solutions for disease detection and treatment. Among different types of nanoparticles (NPs), polymeric nanoparticles comprise biocompatible and biodegradable polymers that enhance drug pharmacokinetics and pharmacodynamics, minimize adverse effects, increase stability, and facilitate sustained drug release. These polymeric nanoparticle-based nanomedicines offer a versatile platform for various cancer treatments, notably enabling targeted drug delivery directly to tumors, tumor-imaging, hyperthermia, and photodynamic therapy. Being polymeric in nature polydopamine (PDA) nanomaterials are appeared as promising approaches in biology and medicine. This review article offers a concise summary of the latest developments in polydopamine-based cancer treatment, covering key findings, limitations, and emerging trend therapeutic approach of polydopamine nanomaterials, along with the properties and various methods of preparation. Physico-chemical properties of PDA-based nanomaterials in therapeutics have permitted several successful modifications in the field of cancer treatment.
Collapse
Affiliation(s)
- Rumi Mahata
- Department of Human Physiology, Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Sounik Manna
- Department of Human Physiology, Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Mrinmoyee Modak
- Department of Human Physiology, Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Sujata Maiti Choudhury
- Department of Human Physiology, Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Vidyasagar University, Midnapore, West Bengal, 721102, India.
| |
Collapse
|
303
|
Li L, Chen M, Reis RL, Kundu SC, Xiao B, Shi X. Advancements of nanoscale drug formulations for combination treatment of colorectal cancer. Int J Pharm 2025; 674:125508. [PMID: 40132771 DOI: 10.1016/j.ijpharm.2025.125508] [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: 01/10/2025] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
Combination chemotherapy is widely utilized in treating colorectal cancer (CRC), particularly for patients who are ineligible for surgery or those with metastatic CRC (mCRC). While this therapeutic method has demonstrated efficacy in managing CRC and mCRC, its broader clinical application is limited due to the unique physical properties, mechanisms of action, and pharmacokinetics of different chemotherapeutic drugs. Consequently, achieving satisfactory treatment outcomes proves to be challenging. Nanotechnology has given rise to innovative drug systems that are precise, controllable, and highly efficient in drug delivery. These nanoscale drug delivery systems can integrate the advantageous aspects of various therapeutic modalities, including chemotherapy, gene therapy, and immunotherapy. This review aims to explain the application of nano-drug delivery system in the treatment of colorectal cancer. Through its unique physical/chemical properties and biological functions, it can solve the limitations of traditional therapy and achieve more accurate, efficient and safe treatment. The advantages/disadvantages, physical and chemical characteristics of various drug delivery systems are described in detail, and suggestions on selecting reasonable NDDSs according to different drug combination methods are given to achieve the best therapeutic effect. This review paper presents an exhaustive summary of the diverse range of drugs utilized in chemotherapy, in addition to outlining strategies for effectively integrating chemotherapy with other treatment modalities. Furthermore, it delves into the principle of selecting carriers for various drug combinations.
Collapse
Affiliation(s)
- Liqi Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Maohua Chen
- Department of Pharmacy, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetic, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4800-058, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetic, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4800-058, Portugal
| | - Bo Xiao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Xiaoxiao Shi
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China.
| |
Collapse
|
304
|
Zhang N, Li J, Ren Y, Xu Y. Comprehensive pan-cancer analysis of PPP1R3G reveals its diagnostic, prognostic, and immunotherapeutic implications. Discov Oncol 2025; 16:530. [PMID: 40232629 PMCID: PMC12000506 DOI: 10.1007/s12672-025-02361-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/10/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND PPP1R3G, a regulatory subunit of protein phosphatase 1, plays a critical role in glycogen metabolism and has been implicated in various cancers. This study provides a comprehensive pan-cancer analysis of PPP1R3G, evaluating its expression, diagnostic and prognostic significance, and potential as a therapeutic target. METHODS We performed an extensive pan-cancer analysis of PPP1R3G using several databases to assess its expression and investigate its correlations with clinical outcomes. Our investigation included assessing PPP1R3G's impact on survival, its correlation with immune checkpoints and tumor stemness scores, and its prognostic significance. We also explored its relationship with immunomodulators, genomic profiles, and immunological characteristics, as well as its response to immunotherapy and involvement in various biological pathways. RESULTS PPP1R3G expression varied significantly across different cancers and correlated with both diagnostic and prognostic outcomes. Moreover, PPP1R3G was significantly linked to immune checkpoints, immunomodulators, prognosis, immunoregulatory genes, tumor stemness, cellular function, and immune infiltration across numerous cancer types. Further analysis of PPP1R3G-related gene enrichment, mutation profiles, RNA modifications, and genomic heterogeneity revealed that missense mutations were the predominant alteration affecting PPP1R3G. CONCLUSIONS Overall, the expression of PPP1R3G is closely associated with various cancers and may serve as a potential biomarker for cancer detection.
Collapse
Affiliation(s)
- Nie Zhang
- Graduate School of Anhui Medical University, Hefei, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Hefei, China
| | - Jiaoyu Li
- Graduate School of Anhui Medical University, Hefei, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Hefei, China
| | - Yanzhi Ren
- Department of Cardiology, Shizhong District People's Hospital, Zaozhuang, China
| | - Yahui Xu
- Graduate School of Anhui Medical University, Hefei, China.
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
305
|
Wei W, Li H, Tian S, Zhang C, Liu J, Tao W, Cai T, Dong Y, Wang C, Lu D, Ai Y, Zhang W, Wang H, Liu K, Fan Y, Gao Y, Huang Q, Ma X, Wang B, Zhang X, Huang Y. Asparagine drives immune evasion in bladder cancer via RIG-I stability and type I IFN signaling. J Clin Invest 2025; 135:e186648. [PMID: 39964752 PMCID: PMC11996873 DOI: 10.1172/jci186648] [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: 09/03/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Tumor cells often employ many ways to restrain type I IFN signaling to evade immune surveillance. However, whether cellular amino acid metabolism regulates this process remains unclear, and its effects on antitumor immunity are relatively unexplored. Here, we found that asparagine inhibited IFN-I signaling and promoted immune escape in bladder cancer. Depletion of asparagine synthetase (ASNS) strongly limited in vivo tumor growth in a CD8+ T cell-dependent manner and boosted immunotherapy efficacy. Moreover, clinically approved L-asparaginase (ASNase),synergized with anti-PD-1 therapy in suppressing tumor growth. Mechanistically, asparagine can directly bind to RIG-I and facilitate CBL-mediated RIG-I degradation, thereby suppressing IFN signaling and antitumor immune responses. Clinically, tumors with higher ASNS expression show decreased responsiveness to immune checkpoint inhibitor therapy. Together, our findings uncover asparagine as a natural metabolite to modulate RIG-I-mediated IFN-I signaling, providing the basis for developing the combinatorial use of ASNase and anti-PD-1 for bladder cancer.
Collapse
Affiliation(s)
- Wenjie Wei
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Hongzhao Li
- Department of Urology, The Third Medical Center and
| | - Shuo Tian
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Chi Zhang
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Junxiao Liu
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Wen Tao
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Tianwei Cai
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Yuhao Dong
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Chuang Wang
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Dingyi Lu
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Yakun Ai
- Department of Pathology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wanlin Zhang
- Department of Pathology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hanfeng Wang
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Kan Liu
- Department of Urology, The Third Medical Center and
| | - Yang Fan
- Department of Urology, The Third Medical Center and
| | - Yu Gao
- Department of Urology, The Third Medical Center and
| | - Qingbo Huang
- Department of Urology, The Third Medical Center and
| | - Xin Ma
- Department of Urology, The Third Medical Center and
| | - Baojun Wang
- Department of Urology, The Third Medical Center and
| | - Xu Zhang
- Department of Urology, The Third Medical Center and
| | - Yan Huang
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
306
|
Keshavarz S, Alavi CE, Aghayan H, Jafari-Shakib R, Vojoudi E. Advancements in Degenerative Disc Disease Treatment: A Regenerative Medicine Approach. Stem Cell Rev Rep 2025:10.1007/s12015-025-10882-z. [PMID: 40232618 DOI: 10.1007/s12015-025-10882-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Regenerative medicine represents a transformative approach to treating nucleus pulposus degeneration and offers hope for patients suffering from chronic low back pain due to disc degeneration. By focusing on restoring the natural structure and function of the nucleus pulposus rather than merely alleviating symptoms, these innovative therapies hold the potential to significantly improve patient outcomes. As research continues to advance in this field, we may soon witness a paradigm shift in how we approach spinal health and degenerative disc disease. The main purpose of this review is to provide an overview of the various regenerative approaches that target the restoration of the nucleus pulposus, a primary site for initiation of intervertebral disc degeneration.
Collapse
Affiliation(s)
- Samaneh Keshavarz
- School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Cyrus Emir Alavi
- Department of Anesthesiology, Neuroscience Research Center, Avicenna University Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Hamidreza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular- Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Jafari-Shakib
- Department of Immunology, School of Medicine, Guilan University of Medical Sciences, P.O.Box 41635 - 3363, Rasht, Iran.
| | - Elham Vojoudi
- Regenerative Medicine, Organ Procurement and Transplantation Multidisciplinary Center, School of Medicine, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
307
|
Yuan J, Hao J, Yu Z, Hu S, Zhai C, Liu J, Cai K. Fabrication of a hierarchical PtIr@Rh hollow trimetallic nanozyme with a higher specific activity than that of HRP for sensitive colorimetric detection. Dalton Trans 2025; 54:6694-6700. [PMID: 40165477 DOI: 10.1039/d5dt00083a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Nanozymes have emerged as promising alternatives to natural enzymes in various fields, owing to their advantages in terms of stability, cost-effectiveness, and multifunctionality. However, their relatively low catalytic activity compared to natural enzymes remains a major challenge for practical applications. Here, we developed hierarchical PtIr@Rh hollow trimetallic nanorods, where Rh served as the substrate and the surface was decorated with numerous Pt nanoparticles doped with a small amount of Ir. The resulting nanorods exhibited remarkable peroxidase-like activity, with a specific activity of 2287 U mg-1, surpassing that of horseradish peroxidase (HRP). Additionally, the maximum reaction velocity (Vmax) was 1.024 × 10-6 M s-1, and the Michaelis-Menten constant (Km) was 1.706 mM. The enhanced catalytic performance was attributed to the unique hierarchical structure and the small amount of Ir doping, as supported by density functional theory (DFT) calculations. The PtIr@Rh nanozyme was successfully applied for the colorimetric detection of L-ascorbic acid, achieving a rapid detection with a limit of detection (LOD) of 0.12 μM. This study introduces a novel nanozyme with superior specific activity compared to natural enzymes, highlighting its potential for colorimetric sensing applications.
Collapse
Affiliation(s)
- Jincheng Yuan
- College of Chemistry & Environmental Engineering, Yangtze University, Jingzhou 434100, China.
| | - Jian Hao
- College of Chemistry & Environmental Engineering, Yangtze University, Jingzhou 434100, China.
| | - Zeyang Yu
- College of Chemistry & Environmental Engineering, Yangtze University, Jingzhou 434100, China.
| | - Shengyang Hu
- College of Chemistry & Environmental Engineering, Yangtze University, Jingzhou 434100, China.
| | - Chenghui Zhai
- College of Chemistry & Environmental Engineering, Yangtze University, Jingzhou 434100, China.
| | - Jiawei Liu
- Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Innovation Academy for Earth Sciences, Chinese Academy of Sciences, Beijing 100029, China.
| | - Kai Cai
- College of Chemistry & Environmental Engineering, Yangtze University, Jingzhou 434100, China.
| |
Collapse
|
308
|
Chang J, Zhang Z, Qu C, Han Q, Xu L. Organic Molecules as a Bridge Connecting Photoelectrochemistry and Fluorescence for Dual-Signal Assay. Anal Chem 2025; 97:7842-7850. [PMID: 40177944 DOI: 10.1021/acs.analchem.4c06431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
We report a strategy based on pyridyl-anchored organic small-molecule fluorescent probes to develop a dual-signal sensing platform. The strategy accomplishes an intelligent integration of fluorescence analysis with photoelectrochemical (PEC) sensing, thereby enabling rapid and precise detection of hypochlorite. In this work, the natural dye chromone was selected as the fluorophore for generating fluorescent signals. Meanwhile, by using phenothiazine (PTZ) as the specific recognition group and pyridine as the anchoring moiety, we designed and synthesized a novel organic small-molecule fluorescent probe. The obtained probe was used as a photosensitive material anchored to the TiO2 surface via N → Ti bonds, to form an FTO/TiO2/FPTZ-1 heterostructure-based dual-signal sensing platform for the detection of hypochlorite. This sensing platform has the characteristics of high specificity, sensitivity, and ease of preparation, enabling rapid qualitative fluorescence readout and quantitative photoelectrochemical readout of hypochlorite, with a limit of detection of 0.288 μM for fluorescence and 1.37 nM for PEC.
Collapse
Affiliation(s)
- Jiaxing Chang
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Zhinan Zhang
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Chulin Qu
- State Key Laboratory of Coordination Chemistry, Collaborative Innovation Center of Advanced Microstructures, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Qingzhi Han
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Li Xu
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
309
|
Li Q, Keskus AG, Wagner J, Izydorczyk MB, Timp W, Sedlazeck FJ, Klein AP, Zook JM, Kolmogorov M, Schatz MC. Unraveling the hidden complexity of cancer through long-read sequencing. Genome Res 2025; 35:599-620. [PMID: 40113261 PMCID: PMC12047254 DOI: 10.1101/gr.280041.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Cancer is fundamentally a disease of the genome, characterized by extensive genomic, transcriptomic, and epigenomic alterations. Most current studies predominantly use short-read sequencing, gene panels, or microarrays to explore these alterations; however, these technologies can systematically miss or misrepresent certain types of alterations, especially structural variants, complex rearrangements, and alterations within repetitive regions. Long-read sequencing is rapidly emerging as a transformative technology for cancer research by providing a comprehensive view across the genome, transcriptome, and epigenome, including the ability to detect alterations that previous technologies have overlooked. In this Perspective, we explore the current applications of long-read sequencing for both germline and somatic cancer analysis. We provide an overview of the computational methodologies tailored to long-read data and highlight key discoveries and resources within cancer genomics that were previously inaccessible with prior technologies. We also address future opportunities and persistent challenges, including the experimental and computational requirements needed to scale to larger sample sizes, the hurdles in sequencing and analyzing complex cancer genomes, and opportunities for leveraging machine learning and artificial intelligence technologies for cancer informatics. We further discuss how the telomere-to-telomere genome and the emerging human pangenome could enhance the resolution of cancer genome analysis, potentially revolutionizing early detection and disease monitoring in patients. Finally, we outline strategies for transitioning long-read sequencing from research applications to routine clinical practice.
Collapse
Affiliation(s)
- Qiuhui Li
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Ayse G Keskus
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Justin Wagner
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - Michal B Izydorczyk
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Texas 77030, USA
- Department of Computer Science, Rice University, Houston, Texas 77251, USA
| | - Alison P Klein
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins Medicine, Baltimore, Maryland 21031, USA
| | - Justin M Zook
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - Mikhail Kolmogorov
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA;
| | - Michael C Schatz
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA;
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins Medicine, Baltimore, Maryland 21031, USA
| |
Collapse
|
310
|
Zeng T, Liao H, Xia L, You S, Huang Y, Zhang J, Liu Y, Liu X, Xie D. Multisite long-read sequencing reveals the early contributions of somatic structural variations to HBV-related hepatocellular carcinoma tumorigenesis. Genome Res 2025; 35:671-685. [PMID: 40037842 PMCID: PMC12047258 DOI: 10.1101/gr.279617.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 01/30/2025] [Indexed: 03/06/2025]
Abstract
Somatic structural variations (SVs) represent a critical category of genomic mutations in hepatocellular carcinoma (HCC). However, the accurate identification of somatic SVs using short-read high-throughput sequencing is challenging. Here, we applied long-read nanopore sequencing and multisite sampling in a cohort of 42 samples from five patients. We found that adjacent nontumor tissue is not entirely normal, as significant somatic SV alterations were detected in these nontumor genomes. The adjacent nontumor tissue is highly similar to tumor tissue in terms of somatic SVs but differs in somatic single-nucleotide variants and copy number variations. The types of SVs in adjacent nontumor and tumor tissue are markedly different, with somatic insertions and deletions identified as early genomic events associated with HCC. Notably, hepatitis B virus (HBV) DNA integration frequently results in the generation of somatic SVs, particularly inducing interchromosomal translocations (TRAs). Although HBV DNA integration into the liver genome occurs randomly, multisite shared HBV-induced SVs are early driving events in the pathogenesis of HCC. Long-read RNA sequencing reveals that some HBV-induced SVs impact cancer-associated genes, with TRAs being capable of inducing the formation of fusion genes. These findings enhance our understanding of somatic SVs in HCC and their role in early tumorigenesis.
Collapse
Affiliation(s)
- Tianfu Zeng
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Haotian Liao
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lin Xia
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Siyao You
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yanqun Huang
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiaxun Zhang
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yahui Liu
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xuyan Liu
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Dan Xie
- Laboratory of Omics Technology and Bioinformatics, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China;
| |
Collapse
|
311
|
Alaridhee ZAI, Alqaraguly MB, Formanova S, Kuryazov R, Mahdi MS, Taher WM, Alwan M, Jabir MS, Zankanah FH, Majdi H, Jawad MJ, Hamad AK, Bozorov K. Recent advances in microfluidic-based photoelectrochemical (PEC) sensing platforms for biomedical applications. Mikrochim Acta 2025; 192:297. [PMID: 40229472 DOI: 10.1007/s00604-025-07135-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/24/2025] [Indexed: 04/16/2025]
Abstract
Photoelectrochemical (PEC) techniques seamlessly combine electrochemical and spectroscopic principles, offering a powerful platform for the detection of biomarkers and biological molecules in clinical and biomedical settings. This review provides a comprehensive overview of microfluidic PEC probes, emphasizing their potential for ultrasensitive detection through enhanced light absorption and charge transfer processes. Key advantages of microfluidic PEC include real-time monitoring of biological processes, non-invasive detection, and the possibility of multiplexing when integrated with various quantification modalities. However, the practical implementation of PEC faces challenges such as bulky setup, matrix interference, and stability of PEC-active materials. Also, this paper discusses the intricate mechanisms of PEC sensing, highlighting the roles of nanomaterials in enhancing microfluidic PEC systems. Additionally, the limitations inherent in PEC material selection, including stability and bandgap engineering, are critically discussed. Solutions such as doping and the development of composite materials are proposed to address these issues. Through presented examples of PEC applications in biomedical fields, this review elucidates the future potential of PEC-based methods as reliable and effective tools for diagnostic applications. Additionally, this review proposes the most effective probes for future investigations to develop commercial devices.
Collapse
Affiliation(s)
| | | | - Shoira Formanova
- Department of Chemistry and Its Teaching Methods, Tashkent State Pedagogical University, Tashkent, Uzbekistan.
| | | | | | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | - Majid S Jabir
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Faeza H Zankanah
- College of Health & Medical Technology, Uruk University, Baghdad, Iraq
| | - Hasan Majdi
- Department of Chemical Engineering and Petroleum Industries, Al-Mustaqbal University College, Babylon, 51001, Iraq
| | | | | | - Khurshed Bozorov
- Department of Organic Synthesis and Bioorganic Chemistry, Institute of Biochemistry, Samarkand State University, University Blvd. 15, 140104, Samarkand, Uzbekistan.
| |
Collapse
|
312
|
Guo C, Lin L, Wang Y, Jing J, Gong Q, Luo K. Nano drug delivery systems for advanced immune checkpoint blockade therapy. Theranostics 2025; 15:5440-5480. [PMID: 40303342 PMCID: PMC12036873 DOI: 10.7150/thno.112475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have been widely utilized in the first-line therapy of various types of cancer. However, immune-related adverse events (irAEs) and resistance to ICIs remain intractable challenges for immune checkpoint blockade (ICB) therapy during clinic treatment. Nano drug delivery systems (NDDSs) have shown promising potential to improve anticancer efficacy and reduce side effects of small molecular drugs. The combination of nanotechnology and ICB provides new opportunities to overcome the challenges of immunotherapy. Nanoplatforms have been employed for direct delivery of ICIs, and they are preferred vehicles for combination therapy of ICIs and other therapeutic agents. In this review, the strategies of using NDDSs for advancing ICB therapy in recent years are surveyed, emphasizing the employment of NDDSs for combination treatment by ICIs and other agents to manipulate antitumor immunity. Analysis of current strategies for applying NDDSs for ICB leads to future research directions and development trends.
Collapse
Affiliation(s)
- Chenqi Guo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ling Lin
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yihan Wang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- West China School of Medicine, Chengdu 610041, China
| | - Jing Jing
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Xiamen Key Lab of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
313
|
Tang S, Yong L, Cui Y, Li H, Bischof E, Cai F. Harnessing Oncolytic Viruses for Targeted Therapy in Triple-Negative Breast Cancer. Int J Med Sci 2025; 22:2186-2207. [PMID: 40303488 PMCID: PMC12035831 DOI: 10.7150/ijms.105683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/19/2025] [Indexed: 05/02/2025] Open
Abstract
Breast cancer is the most prevalent malignant tumor among women, with triple-negative breast cancer (TNBC) being one of the most aggressive forms due to its high invasiveness and metastatic potential. Traditional treatments such as endocrine therapy and anti-HER2-targeted therapy are largely ineffective for TNBC, and while chemotherapy shows some promise, resistance remains a significant hurdle. Recently, there has been increasing interest in biological therapies, especially oncolytic viruses (OVs). OVs promote anti-tumor effects by selectively killing tumor cells and stimulating immune responses, and have achieved notable breakthroughs in breast cancer treatment. OVs have demonstrated effectiveness comparable to surgery, radiotherapy, or chemotherapy in selected cancers, but data are sparse in the context of TNBC. This review provides an overview of recent progress in the application of OVs as a tool for precision TNBC treatment.
Collapse
Affiliation(s)
- Shasha Tang
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai 200065, China
| | - Liyun Yong
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai 200065, China
| | - Yong Cui
- Department of General Surgery, People's Hospital of Otog Qianqi, Sharita Tara East Street, Aolezhaoqi Town, Otog Qianqi 016200, China
| | - Haibin Li
- Department of General Surgery, People's Hospital of Otog Qianqi, Sharita Tara East Street, Aolezhaoqi Town, Otog Qianqi 016200, China
| | - Evelyne Bischof
- Department of Medical Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Fengfeng Cai
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd, Shanghai 200065, China
| |
Collapse
|
314
|
Ramírez-Sánchez A, Jociles-Ortega M, García-Martinez JM, Torrens-Martínez I, Martínez-Useros J, Fernández-Aceñero MJ, Blas TOD, Louphrasitthiphol P, Goding CR, García-Jiménez C, Chocarro-Calvo A. Metabolic buffering suppresses phenotype switching in cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.647576. [PMID: 40291724 PMCID: PMC12026899 DOI: 10.1101/2025.04.07.647576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The impact of the microenvironment on epigenetically plastic cancer cells underpins phenotypic heterogeneity, a major cause of metastatic dissemination and therapy resistance that together represent the primary cause of cancer-related death. Nutrient limitation is a key microenvironmental stress that can cause a phenotypic transition from proliferation to invasion via activation of the integrated stress response. However, whether and how the capacity to store and mobilize nutrients impacts phenotype-switching through metabolic buffering remains unknown. Here, using melanoma as a model, we reveal that the ability to accumulate and mobilize glycogen, that buffers glucose availability, plays a key role in phenotypic transitions in melanoma. While proliferative phenotype cells exhibit high levels of glycogen, invasion is marked by low glycogen levels. Significantly, an inability to store and metabolize glycogen leads to phenotype instability and a switch to invasion. Accordingly, glycogen levels inversely correlate with Clark levels in primary melanomas, with low expression of the glycogen phosphorylases PYGB/L and phosphoglucomutase 1 (PGM1) being associated with worse overall survival. The importance of metabolic buffering in suppressing phenotypic transitions likely extrapolates to other cancer types. Highlights Melanoma phenotypes are distinguished by their ability to store and mobilize glycogen. Proliferative MITF High melanoma cells store glycogen to improve survival under stressful conditions. Inhibition of glycogen degradation impairs proliferation in MITF High melanoma cells. Lack of PGM1 drives invasion and metastatic dissemination.
Collapse
|
315
|
Guo X, Zhao Z, Zhu L, Liu S, Zhou L, Wu F, Fang S, Chen M, Zheng L, Ji J. The evolving landscape of biomarkers for systemic therapy in advanced hepatocellular carcinoma. Biomark Res 2025; 13:60. [PMID: 40221793 PMCID: PMC11993949 DOI: 10.1186/s40364-025-00774-2] [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: 01/26/2025] [Accepted: 03/29/2025] [Indexed: 04/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains one of the most prevalent and deadliest cancers. With the approval of multiple first- and second-line agents, especially the combination therapies based on immune checkpoint inhibitor (ICI) regimens, the landscape of systemic therapy for advanced HCC (aHCC) is more diverse than ever before. The efficacy of current systemic therapies shows great heterogeneity in patients with aHCC, thereby identifying biomarkers for response prediction and patient stratification has become an urgent need. The main biomarkers for systemic therapy in hepatocellular carcinoma are derived from peripheral blood, tissues, and imaging. Currently, the understanding of the clinical response to systemic therapy indicates unequivocally that a single biomarker cannot be used to identify patients who are likely to benefit from these treatments. In this review, we provide an integrated landscape of the recent development in molecular targeted therapies and ICIs-based therapies, especially focusing on the role of clinically applicable predictive biomarkers. Additionally, we further highlight the latest advancements in biomarker-driven therapies, including targeted treatments, adoptive cell therapies, and bispecific antibodies.
Collapse
Affiliation(s)
- Xinyu Guo
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Zhongwei Zhao
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Lingyi Zhu
- The 2nd Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, China
| | - Shuang Liu
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Lingling Zhou
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Fazong Wu
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Shiji Fang
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Minjiang Chen
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Liyun Zheng
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China.
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
| | - Jiansong Ji
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China.
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
| |
Collapse
|
316
|
Nunes ADC, Pitcher LE, Exner HA, Grassi DJ, Burns B, Sanchez MBH, Tetta C, Camussi G, Robbins PD. Attenuation of Cellular Senescence and Improvement of Osteogenic Differentiation Capacity of Human Liver Stem Cells Using Specific Senomorphic and Senolytic Agents. Stem Cell Rev Rep 2025:10.1007/s12015-025-10876-x. [PMID: 40220121 DOI: 10.1007/s12015-025-10876-x] [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] [Accepted: 04/02/2025] [Indexed: 04/14/2025]
Abstract
Expansion of adult stem cells in culture increases the percent of senescent cells, reduces their differentiation capacity and limits their clinical use. Here, we investigated whether treatment with certain senotherapeutic drugs would reduce the accumulation of senescent cells during expansion of human liver stem cells (HLSCs) while maintaining their differentiation capacity. Our results demonstrate that chronic treatment with the senomorphic XJB-5-131 or the senolytics cocktail D + Q reduced the number of senescent cells and significantly reduced the expression of senescence-associated genes and several inflammatory SASP factors in later passage HLSCs. Additionally, treatment with XJB-5-131 and D + Q improved the capacity of HLSCs to undergo osteogenic differentiation following extensive in vitro expansion. Overall, our data demonstrate that treatment with XJB-5-13 or D + Q results in a reduction in the percentage of replication-induced senescent HLSCs and likely other types of adult stem cells and improve the potential therapeutic use of later passage human stem cells.
Collapse
Affiliation(s)
- Allancer D C Nunes
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| | - Louise E Pitcher
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| | - Henry A Exner
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Brittan Burns
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| | - Maria Beatriz Herrera Sanchez
- Molecular Biotechnology Centre, University of Torino, Torino, Italy
- 2i3T Societ Per la Gestione Dell'incubatore di Imprese e per il Trasferimento Tecnologico Scarl, University of Torino, Torino, Italy
| | | | - Giovanni Camussi
- Molecular Biotechnology Centre, University of Torino, Torino, Italy
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Paul D Robbins
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
317
|
Liang HY, Luo RZ, Deng R, Chen SL, Liu X, Yang X, Wei LJ, Wei ZQ, Wu LY, Shen HM, Yun JP, Liu LL. Glycogen stores mediated by the p53-GYS1 feedback circuit engenders platinum resistance in ovarian clear cell carcinoma. Cell Death Differ 2025:10.1038/s41418-025-01500-z. [PMID: 40210982 DOI: 10.1038/s41418-025-01500-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 02/27/2025] [Accepted: 03/21/2025] [Indexed: 04/12/2025] Open
Abstract
Ovarian cancer (OC) is a highly fatal and refractory malignancy affecting women, and platinum resistance remains a major clinical dilemma. Compared with other OC subtypes, ovarian clear cell carcinoma (OCCC) frequently exhibits increased platinum refractoriness, accompanied by increased glycogen levels, which promotes clear-cell morphology, and wild-type p53. However, the roles of these factors in platinum resistance of OCCC are unclear. Here, we investigated whether glycogen promotes OCCC resistance to platinum agents and reported that GYS1, a rate-limiting enzyme in glycogen synthesis, is clinically associated with poor prognosis and chemoresistance in OCCC. Mechanistically, p53 promotes GYS1 breakdown via the upregulation of RNF144a, whereas GYS1 induces the reversal of p53 ubiquitination and degradation by competitively binding to USP14, forming a positive feedback circuit. Under platinum stress, the accumulated glycogen is mobilized by the p53/GYS1 feedback circuit, which fuels energetic NADPH production, resulting in resistance to disulfidptosis and increased platinum resistance in OCCC. Collectively, our findings identify glycogen as a contributor to OCCC platinum resistance and elucidate the underlying mechanisms, highlighting a crucial p53/GYS1 positive feedback loop.
Collapse
Affiliation(s)
- Hao-Yu Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Rong-Zhen Luo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Ru Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Shi-Lu Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xuan Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xia Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Li-Jun Wei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Zong-Qiang Wei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Li-Yan Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Hui-Min Shen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China.
| | - Jing-Ping Yun
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Li-Li Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
318
|
Justiz-Vaillant A, Pandit BR, Unakal C, Vuma S, Akpaka PE. A Comprehensive Review About the Use of Monoclonal Antibodies in Cancer Therapy. Antibodies (Basel) 2025; 14:35. [PMID: 40265416 PMCID: PMC12015915 DOI: 10.3390/antib14020035] [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: 01/27/2025] [Revised: 03/14/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Monoclonal antibodies (mAbs) targeting various pathways in cancer therapy play crucial roles in enhancing the immune system's ability to recognise and eliminate tumour cells. These therapies are designed to either block inhibitory immune checkpoint pathways or to target specific tumour cell markers for direct destruction. Additionally, mAbs can modulate the tumour microenvironment, enhance antibody-dependent cellular cytotoxicity, and inhibit angiogenesis, further amplifying their therapeutic impact. Below is a summary of monoclonal antibodies targeting key pathways, along with their indications and mechanisms of action, which are reviewed based on therapeutic mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Patrick Eberechi Akpaka
- Department of Pathology/Microbiology & Pharmacology, The University of the West Indies, St. Augustine Campus, St. Augustine 330912, Trinidad and Tobago; (A.J.-V.); (B.R.P.); (C.U.); (S.V.)
| |
Collapse
|
319
|
Rabiee N, Rabiee M. Engineered Metal-Organic Frameworks for Targeted CRISPR/Cas9 Gene Editing. ACS Pharmacol Transl Sci 2025; 8:1028-1049. [PMID: 40242591 PMCID: PMC11997888 DOI: 10.1021/acsptsci.5c00047] [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: 01/16/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 04/18/2025]
Abstract
The development of precise and efficient delivery systems is pivotal for advancing CRISPR/Cas9 gene-editing technologies, particularly for therapeutic applications. Engineered metal-organic frameworks (MOFs) have emerged as a promising class of inorganic nonviral vectors, offering unique advantages such as tunable porosity, high cargo-loading capacity, and biocompatibility. This review explores the design and application of MOF-based nanoplatforms tailored for the targeted delivery of CRISPR/Cas9 components, aiming to enhance gene-editing precision and efficiency. By incorporating stimuli-responsive linkers and bioactive ligands, these MOFs enable controlled release of CRISPR/Cas9 payloads at the target site. Comparative discussions demonstrate superior performance of MOFs over conventional nonviral systems in terms of stability, transfection efficiency, and reduced off-target effects. Additionally, the intracellular trafficking mechanisms and the therapeutic potential of these platforms in preclinical models are discussed. These findings highlight the transformative potential of MOF-based delivery systems in overcoming the challenges associated with gene-editing technologies, such as immunogenicity and cytotoxicity, paving the way for their application in precision medicine. This review provides a blueprint for the integration of nanotechnology and genome editing, advancing the frontier of nonviral therapeutic delivery systems.
Collapse
Affiliation(s)
- Navid Rabiee
- Department
of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua−Peking
Joint Center for Life Sciences, Tsinghua
University, Beijing 100084, China
- MOE
Key Laboratory of Bioinformatics, Tsinghua
University, Beijing 100084, China
- Department
of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India
| | - Mohammad Rabiee
- Biomaterials
Group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran 165543, Iran
| |
Collapse
|
320
|
Li Z, Yi H, Li Y, Yang J, Guo P, Han F. Identification and validation of a novel autophagy-related biomarker in obstructive sleep apnea syndrome. Sleep 2025; 48:zsae287. [PMID: 39665515 DOI: 10.1093/sleep/zsae287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/24/2024] [Indexed: 12/13/2024] Open
Abstract
STUDY OBJECTIVES Obstructive sleep apnea syndrome is closely associated with tumor growth. Chronic intermittent hypoxia promotes autophagy and is related to malignant tumor development. However, the role of autophagy in obstructive sleep apnea syndrome progression remains unclear. METHODS obstructive sleep apnea syndrome datasets (GSE135917 and GSE38792) from Gene Expression Omnibus were analyzed to identify differentially expressed genes and autophagy-related differentially expressed genes. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and gene set enrichment analysis were conducted, and a protein-protein interaction network identified hub genes. Colorectal cancer datasets from The Cancer Genome Atlas were used for differential expression and survival analyses, along with gene set enrichment analysis and immune infiltration analysis. Chronic intermittent hypoxia-induced autophagy and oxidative stress were investigated in Sprague-Dawley rats using reactive oxygen species assays. Hub genes were validated in rats and obstructive sleep apnea syndrome patient samples. RESULTS Gene set enrichment analysis revealed significant differences in autophagy-related gene expression among obstructive sleep apnea syndrome patients. Hub genes ATG5, CASP1, MAPK8, EIF4G1, and TANK-binding kinase 1 were identified, with ATG5 and TANK-binding kinase 1 validated. Autophagy-related differentially expressed genes were predominantly upregulated in colorectal cancer tissues. TANK-binding kinase 1 expression in colorectal cancer patients was associated with enhanced sensitivity to immunotherapy and CD8 + T cell, macrophage, and regulatory T cell infiltration, potentially influencing the immune microenvironment. The animal experiments showed that chronic intermittent hypoxia increased reactive oxygen species levels, suggesting that chronic intermittent hypoxia plays a role in autophagy. TANK-binding kinase 1 expression was significantly higher in obstructive sleep apnea syndrome patients than in controls, and continuous positive airway pressure did not alter TANK-binding kinase 1 levels. CONCLUSIONS This study is the first to describe the potential contribution of TANK-binding kinase 1 to the development of obstructive sleep apnea syndrome and its potential as a novel biomarker and potential therapeutic target for obstructive sleep apnea syndrome.
Collapse
Affiliation(s)
- Zhiyong Li
- Department of Emergency Surgery, Peking University People's Hospital, Xicheng, Beijing, China
| | - Huijie Yi
- Department of Sleep Medicine, Peking University People's Hospital, Xicheng, Beijing, China
| | - Yuxi Li
- Department of Emergency Surgery, Peking University People's Hospital, Xicheng, Beijing, China
| | - Jie Yang
- Department of Emergency Surgery, Peking University People's Hospital, Xicheng, Beijing, China
| | - Peng Guo
- Department of Emergency Surgery, Peking University People's Hospital, Xicheng, Beijing, China
| | - Fang Han
- Department of Sleep Medicine, Peking University People's Hospital, Xicheng, Beijing, China
| |
Collapse
|
321
|
Zhao H, Chen W, Zhu Y, Chao Z, Sun J, Zhang Q, Guo H, Ju H, Liu Y. Programming a multiplex lanthanide nanoparticle for customized cancer treatment with real-time efficiency feedback. NANOSCALE 2025; 17:9184-9196. [PMID: 40125577 DOI: 10.1039/d5nr00390c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Customized cancer therapy relies on timely therapeutic effect evaluation to provide prescription adjustment for individual cases. However, currently reported therapeutic reagents are rarely integrated with imaging probes for self-evaluation of effects. Contrast imaging agents to measure tumor size changes must be administrated separately after therapy, complicating the therapeutic process and delaying reporting time. Herein, we design a customized therapy platform (LNPs-RB/Pep/cRGD) by conjugating lanthanide nanoparticles (LNPs) with the photosensitizer rose bengal, a caspase-3 substrate peptide (with Cy7.5 labelled at the terminal), and the tumor-targeting molecule cRGD. LNPs exhibit NIR-IIb downconversion luminescence under 980 nm/808 nm excitations for in vivo imaging, and visible upconversion luminescence under high-power 980 nm excitation for photodynamic therapy (PDT). By sequentially programming NIR excitation wavelength and power, NIR-IIb-imaging guided PDT and real-time cancer cell apoptosis imaging are achieved as therapeutic efficiency feedback. PDT induces cell apoptosis, generating caspase-3, which cleaves Cy7.5-containing peptide fragments from LNPs. This process corresponds to a recovery in vivo of NIR-IIb ratiometric imaging at 808 nm versus 980 nm excitation. The cleaved Cy7.5-containing peptide fragment is cleared into urine for NIR imaging. Both cell apoptosis imaging processes are completed 12 h after PDT, which is 7 days earlier than tumor size measurement. Therefore, customized therapy is achieved by timely adjusting PDT dosage, enhancing therapeutic efficacy.
Collapse
Affiliation(s)
- Hongxia Zhao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Jiangsu, Taizhou 225300, China
| | - Wei Chen
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nan-jing University, Nanjing 210023, China
| | - Yu Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Zhicong Chao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Jiahui Sun
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Qing Zhang
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nan-jing University, Nanjing 210023, China
| | - Hongqian Guo
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nan-jing University, Nanjing 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Ying Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| |
Collapse
|
322
|
Gao X, Xu Y, Hu X, Chen J, Zhang D, Xu X. Comprehensive analysis of mitochondrial solute carrier family 25 (SLC25) identifies member 19 (SLC25A19) as a regulatory factor in hepatocellular carcinoma. Gene 2025; 944:149299. [PMID: 39892835 DOI: 10.1016/j.gene.2025.149299] [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: 11/01/2024] [Revised: 01/08/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND The mitochondrial solute carrier family 25 (SLC25) is known to play a pivotal role in oncogenesis, yet its specific involvement in hepatocellular carcinoma (HCC) remains poorly elucidated. METHODS In this study, we performed a clustering analysis of HCC patients in the Cancer Genome Atlas database based on the expression levels of SLC25 members, and conducted clinical feature analysis for each patient within the clusters. Subsequently, we developed a prognostic model using a Lasso regression approach with SLC25A19, SLC25A49, and SLC25A51 as features, and generated a risk score for each HCC patient. We then identified SLC25A19 as a potential prognostic marker for HCC through single-cell analysis, and validated this finding using in vitro and in vivo experiments. RESULTS Our results revealed significant differences in the expression of most SLC25 family members in HCC patients, enabling the stratification of patients into three clusters, with those in cluster 1 exhibiting the most favorable prognosis and showing a correlation with enhanced immune infiltration. The risk scores derived from the features SLC25A19, SLC25A49, and SLC25A51 effectively predicted the prognosis of HCC patients, with area under the curve (AUC) values exceeding 0.7 in the test group. Single-cell analysis further demonstrated h eightened expression of SLC25A19 in the immune microenvironment of HCC, and in vitro experiments indicated that SLC25A19 may regulate the proliferation, migration, invasion, cycle, and apoptosis of liver cancer cells through the Wnt pathway. In the HepG2 animal model, overexpression of SLC25A19 significantly promotes tumor growth, while knockdown inhibits tumor growth. Analysis of patient tumor tissues shows that SLC25A19 is highly expressed in liver cancer tissues and is associated with CD8+ T cell infiltration. CONCLUSIONS In conclusion, our comprehensive analysis of the role of SLC25 in HCC unveiled SLC25A19 as a potential regulatory factor in HCC.
Collapse
Affiliation(s)
- Xueke Gao
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Yangtao Xu
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Xinyao Hu
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Jiayu Chen
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Daoming Zhang
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Ximing Xu
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060.
| |
Collapse
|
323
|
Li Y, Sun Q, Yang Z, Luo M, Yang W, Song Z, Liu K, Li H, Gao W, Wu Q, Shen W, Yang Y, Yin D. Transforming tumors into 'high-risk bombs' triggers a neoantigen storm and amplifies immune responses. J Control Release 2025; 380:1080-1094. [PMID: 39971250 DOI: 10.1016/j.jconrel.2025.02.040] [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: 11/11/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
Although various immunotherapies have improved the treatment of several challenging malignancies in clinical applications, current research suggests that neoantigens remain fundamental to the initiation of immunotherapy, implying a dependence on high mutation loads in tumors and stable target antigens. To overcome these limitations, we propose a novel immunotherapy paradigm that interferes with splicing to induce the expression of neoantigens and neoepitopes while simultaneously blocking autophagy to prevent their degradation through endogenous pathways. This approach ensures the stable expression and accumulation of neoantigens and neoepitopes in tumor cells. To fully unleash the potential of neoantigens, we further induce tumors to undergo immunogenic cell death (ICD), triggering a "neoantigen storm" at the tumor site to recruit and activate more dendritic cells (DCs). Through a DC-dependent mechanism, communication between the tumor and the tumor-draining lymph node (TDLN) is enhanced, summoning more neoantigen-specific cytotoxic T lymphocytes to lyse tumor cells and establish immune circulation. In summary, this work presents a novel antigen-based immune sensitization strategy that stabilizes target antigens while exploring the potential of non-targeted antigens. By bypassing the cumbersome neoantigen identification process, this strategy holds promise for rapid clinical application in combination with other immunotherapies.
Collapse
Affiliation(s)
- Yunlong Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Quanwei Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Zexin Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Min Luo
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wenshuo Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Zhengwei Song
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Kang Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Huihui Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wenheng Gao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Qinghua Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China; Anhui Provincial Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230021, China.
| | - Ye Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230031, China; Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China.
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China; Anhui Provincial Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230021, China; Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China.
| |
Collapse
|
324
|
Yao Y, Li Y, Hua Q, Zhao W, Li J. Development of novel poly(1,4-benzenedithiol) nanoparticles@AuNPs lateral flow immunochromatographic test strips. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:2997-3006. [PMID: 40160149 DOI: 10.1039/d5ay00346f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
In this study, we successfully developed a novel lateral flow immunochromatography (LFIA) strip that utilizes poly(1,4-benzenedithiol) (PBDT) nanoparticles loaded with gold nanoparticles (PBDT@AuNPs), characterized by a particle size of 147 nm and a spherical morphology, as labeling materials to enhance the sensitivity of colloidal gold. PBDT@AuNPs exhibit a higher loading capacity and molar extinction coefficient compared to traditional markers, with an approximately 22-fold increase in molar extinction coefficient relative to colloidal gold immunochromatography, significantly improving detection sensitivity. Cardiac troponin I (cTnI) was employed as a model to assess the detection efficiency and reliability of PBDT@AuNPs LFIA strips. A calibration curve was established using a four-parameter logistic function to fit the relationship between colloidal gold colorimetric strength and cTnI concentration, covering a range of 0.06 to 500 ng mL-1, with a detection limit of 0.03 ng mL-1 as calculated by the International Union of Pure and Applied Chemistry (IUPAC). A chemical comparison of various cTnI concentrations in human serum samples, conducted using Passing-Bablok regression, confirmed the equivalence of the PBDT@AuNPs LFIA strips with a commercial fluorescent cTnI LFIA kit, revealing no significant systematic or proportional bias. The spike recovery ranged from 91.92% to 108.70%, and the relative standard deviation (RSD) ranged from 5.29% to 5.85%, underscoring the high precision of the PBDT@AuNPs LFIA strips in quantifying cTnI in human serum samples.
Collapse
Affiliation(s)
- Yu Yao
- Guangxi Key Laboratory of Electrochemical and Magneto-Chemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin, 541004, China.
| | - Yafei Li
- Guangxi Key Laboratory of Electrochemical and Magneto-Chemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin, 541004, China.
| | - Qingqing Hua
- Guangxi Key Laboratory of Electrochemical and Magneto-Chemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin, 541004, China.
| | - Wenlin Zhao
- Guangxi Key Laboratory of Electrochemical and Magneto-Chemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin, 541004, China.
| | - Jishun Li
- Guangxi Key Laboratory of Electrochemical and Magneto-Chemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin, 541004, China.
- College of Environmental Science and Engineering, Guilin University of Technology, Guilin, 541004, China
| |
Collapse
|
325
|
Xu Y, Yang S, Rao Q, Gao Y, Zhou G, Zhao D, Shi X, Chai Y, Zhao C. A mechanistic quantitative systems pharmacology model platform for translational efficacy evaluation and checkpoint combination design of bispecific immuno-modulatory antibodies. Front Pharmacol 2025; 16:1571844. [PMID: 40276607 PMCID: PMC12018249 DOI: 10.3389/fphar.2025.1571844] [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: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Over the past 2 decades, tumor immunotherapies have witnessed remarkable advancements, especially with the emergence of immune checkpoint-targeting bispecific antibodies. However, a quantitative understanding of the dynamic cross-talking mechanisms underlying different immune checkpoints as well as the optimal dosing and target design of checkpoint-targeting bispecific antibodies still remain challenging to researchers. To address this challenge, we have here developed a multi-scale quantitative systems pharmacology (QSP) model platform that integrates a diverse array of immune checkpoints and their interactive functions. The model has been calibrated and validated against an extensive collection of multiscale experimental datasets covering 20+ different monoclonal and bispecific antibody treatments at over 60 administered dose levels. Based on high-throughput simulations, the QSP model platform comprehensively screened and characterized the potential efficacy of different bispecific antibody target combination designs, and model-based preclinical population-level simulations revealed target-specific dose-response relationships as well as alternative dosing strategies that can maintain anti-tumor treatment efficacy while reducing dosing frequencies. Model simulations also pointed out that combining checkpoint-targeting bispecific antibodies with monoclonal antibodies can lead to significantly enhanced anti-tumor efficacy. Our mechanistic QSP model can serve as an integrated precision medicine simulation platform to guide the translational research and clinical development of checkpoint-targeting immuno-modulatory bispecific antibodies.
Collapse
Affiliation(s)
- Yiyang Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Siyuan Yang
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Qi Rao
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yuan Gao
- QSPMed Technologies, Nanjing, China
| | - Guanyue Zhou
- Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing, China
| | - Dongmei Zhao
- Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing, China
| | - Xinsheng Shi
- Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing, China
| | - Yi Chai
- Phase I Clinical Trial Unit, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Zhao
- School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
326
|
Liu S, Ji Z, Ge S, Cai C, Zhang H, Wang Y, Zhou Y, Zhou J, Cheng H, Ding Y. Cascade-Targeting Apoptosis via Synergy of TRAIL-Specific Bystander Effect and Mitochondrial Photodamage in Cancer Therapy. NANO LETTERS 2025; 25:5881-5890. [PMID: 40151999 DOI: 10.1021/acs.nanolett.5c00878] [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: 03/29/2025]
Abstract
Tumor-specific apoptosis exerts considerable curative efficacy in cancer, particularly with TRAIL, which has been approved in the clinic; however, therapeutic outcome is compromised due to apoptosis evasion and the short half-life of exogenously infused TRAIL. Herein, we propose a synergistic apoptosis strategy of orthotopic TRAIL expression for enhancing the bystander effect and mitochondrial photodamage for intrinsic apoptosis activation. To actualize synergetic apoptosis, we develop cascade-targeting nanoparticles to perform cell-to-mitochondria shuttling, in which TRAIL-expressing plasmid (pTRAIL) is coprecipitated with calcium phosphate on a glycyrrhetinic acid (GA)-modified graphene oxide nanosheet. For apoptosis synergy, GA mediates tumor accumulation of nanoparticles, followed by structure dissociation for efficient pTRAIL release and expression (cascade module I). Thereafter, GA-modified graphene carriers perform mitochondria distribution for laser-triggered photodamage (cascade module II). The nanoparticles yield tumor inhibition of 86.78% in the melanoma model and demonstrate metastasis blocking activity. Collectively, a cascade-targeting apoptosis technology via a combination of TRAIL-specific bystander effects and mitochondrial photodamage provides innovative oncotherapy synergy.
Collapse
Affiliation(s)
- Shengyu Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Zhongsheng Ji
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Sulei Ge
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Chuxin Cai
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Huaqing Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yifan Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Cheng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Ding
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
| |
Collapse
|
327
|
Holca A, Cucuiet V, Astilean S, Lamy de la Chapelle M, Focsan M. Recent advances in gold nanoparticle-graphene hybrid nanoplatforms with visible to near-infrared response for photodynamic and photothermal therapy and bioimaging. RSC Adv 2025; 15:11902-11922. [PMID: 40236567 PMCID: PMC11998979 DOI: 10.1039/d4ra09100k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/23/2025] [Indexed: 04/17/2025] Open
Abstract
Photodynamic therapy (PDT) and photothermal therapy (PTT) are light-activated cancer treatments. PDT involves the administration of a photosensitizing agent, which is activated by light of a specific wavelength to generate reactive oxygen species. Alternatively, PTT involves the use of photothermal agents, which are materials that absorb light and convert it into heat. Gold nanoparticles are often used as photothermal agents owing to their localized surface plasmon resonance (LSPR), a key optical property, which allows them to efficiently absorb light and convert it into heat. Graphene, which is a 2D material with extraordinary optical and physical properties and a large surface area, shows great promise both in PDT and PTT as an intrinsic nanoheater or a versatile platform for the immobilization of gold nanoparticles and other functional molecules, including photosensitizers. Moreover, graphene-based derivatives, i.e. graphene oxide (GO) and reduced graphene oxide (rGO), exhibit intrinsic optical/spectroscopic signals, which can be used in fluorescence, Raman and thermal imaging. By combining gold nanoparticles with graphene derivatives, a higher increase in temperature can be achieved under light irradiation owing to the synergistic effect of these two materials and the drug delivery efficiency and multimodal imaging techniques can be enhanced. This review provides insights into graphene-based nanoplatforms, focusing on multimodal therapy and imaging techniques. Furthermore, future perspectives in the field of graphene-based- and hybrid-nanoplatforms are suggested.
Collapse
Affiliation(s)
- Alexandru Holca
- Department of Biomolecular Physics, Faculty of Physics, Babes-Bolyai University M. Kogalniceanu 1 400084 Cluj-Napoca Romania
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University T. Laurian 42 400271 Cluj-Napoca Romania
| | - Vlad Cucuiet
- Department of Biomolecular Physics, Faculty of Physics, Babes-Bolyai University M. Kogalniceanu 1 400084 Cluj-Napoca Romania
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University T. Laurian 42 400271 Cluj-Napoca Romania
| | - Simion Astilean
- Department of Biomolecular Physics, Faculty of Physics, Babes-Bolyai University M. Kogalniceanu 1 400084 Cluj-Napoca Romania
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University T. Laurian 42 400271 Cluj-Napoca Romania
| | - Marc Lamy de la Chapelle
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University T. Laurian 42 400271 Cluj-Napoca Romania
- Le Mans Institute of Molecules and Materials (IMMM - UMR6283), Le Mans University Avenue Olivier Messiaen Le Mans 72085 Cedex 9 France
| | - Monica Focsan
- Department of Biomolecular Physics, Faculty of Physics, Babes-Bolyai University M. Kogalniceanu 1 400084 Cluj-Napoca Romania
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University T. Laurian 42 400271 Cluj-Napoca Romania
| |
Collapse
|
328
|
Li Q, Chen Q, Xiao S, Wang S, Ge X, Wang Q, Zheng L, Wei Q, Du W, Shen W, Wu Y, Song J. A Salidroside-Based Radiosensitizer Regulates the Nrf2/ROS Pathway for X-Ray Activated Synergistic Cancer Precise Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2413226. [PMID: 40195850 DOI: 10.1002/adma.202413226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/26/2025] [Indexed: 04/09/2025]
Abstract
The hypoxic microenvironment and radioresistance of tumor cells, as well as the delay in efficacy evaluation, significantly limit the effect of clinical radiotherapy. Therefore, developing effective radiosensitizers with monitoring of tumor response is of great significance for precise radiotherapy. Herein, a novel radiosensitizer (term as: SCuFs) is developed, consisting of traditional Chinese medicine (TCM) compounds salidroside, Cu2+, and hydroxyl radical (•OH) activated second near-infrared window fluorescence (NIR-II FL) molecules, which make the radiosensitization effect and boosted chemodynamic therapy (CDT) efficacy. The overexpressed glutathione in the tumor induces the SCuFs dissociation, allowing deep penetration of the drug to the whole tumor region. After X-ray irradiation, salidroside inhibits the Nuclear factor erythroid 2-like 2 (Nrf2)protein expression and blocks cells in the G2/M phase with the highest radiosensitivity, which amplifies the reactive oxygen species (ROS) generation to exacerbate DNA damage, thus achieving radiosensitization. Meanwhile, the upregulated ROS provides sufficient chemical fuel for Cu+-mediated CDT to produce more •OH. NIR-II FL imaging can monitor the •OH changes during the therapy process, confirming the radiosensitization effect and CDT process related to •OH. This study not only achieves effective radiosensitization and cascaded ROS-mediated CDT efficacy, but also provides a useful tool for monitoring therapeutic efficacy, showing great prospects for clinical application.
Collapse
Affiliation(s)
- Qingqing Li
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Qing Chen
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Shenggan Xiao
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Shuhan Wang
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Xiaoguang Ge
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Qian Wang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Liting Zheng
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Qiaoqiao Wei
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Wei Du
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Wenbin Shen
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Ying Wu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
329
|
Lu FL, Wu YY, Feng SJ, Yang XS, Tian X, Ma HY, Wang W, Zhang WS, Han DX, Qin DD, Han DF, Niu L. Signal "Off-On Model'' for Colorimetric Sensing Proanthocyanidin B 2 Achieved by the Organic Solvent-Processed Amorphous BiVO 4 Nanozyme. Anal Chem 2025; 97:7165-7176. [PMID: 40131118 DOI: 10.1021/acs.analchem.4c06509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Here, we report a "off-on model"-based colorimetric sensor without the assistance of H2O2, achieved by organic solvent-processed amorphous BiVO4 nanoparticles favorable for large-scale manufacturing. The amorphous material beyond traditional crystalline nanozymes has both vanadium vacancy (Vv) and oxygen vacancy (Ov), as well as single oxidase-like activity with both hydroxyl radical (•OH) and superoxide anion (O2•-) as electron acceptors. Despite the high enzymatic performance, the dual-defect-rich amorphous BiVO4 preserves good long-term activity in either buffer solution or the solid state. The specially designed BiVO4 is capable of accelerating TMB oxidation in the presence of polyphenols, thus leading to an interesting signal-intensified colorimetric response. This is probably due to the coordination of the ortho dihydroxyl group in polyphenols with the catalyst by replacing the surface-adsorbed NO3-, which results in faster charge transfer between BiVO4 and substrate and, thus, more rapid depletion of radicals for TMB oxidation. Based on these findings, a sensor platform for proanthocyanidin (PAC) B2 is established with a limit of detection (LOD) as low as 65 nM and good specificity. The sensor shows higher sensitivity than the standard Porter method (LOD: 0.29 μM) and comparable accuracy when analyzing PAC B2 in commercially available grape seed capsules, with quantitative recoveries varying from 104.0 to 108.3% and relative standard deviations ranging from 1.7 to 5.7%. To date, this is the only nanozyme-based chemical sensor that is active for the signal "off-on model" for the detection of reducing polyphenols. Also, this method seems quite general, and it can be used for sensing of a series of specific polyphenols in addition to PAC B2.
Collapse
Affiliation(s)
- Fu-Long Lu
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Laboratory of Optoelectronic Materials and Sensor Components, Guangzhou Key Laboratory of Sensing Materials & Devices, Centre for Advanced Analytical Science, Guangzhou University, Guangzhou 510006, P. R. China
| | - Yun-Yun Wu
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Laboratory of Optoelectronic Materials and Sensor Components, Guangzhou Key Laboratory of Sensing Materials & Devices, Centre for Advanced Analytical Science, Guangzhou University, Guangzhou 510006, P. R. China
| | - Shu-Jie Feng
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Laboratory of Optoelectronic Materials and Sensor Components, Guangzhou Key Laboratory of Sensing Materials & Devices, Centre for Advanced Analytical Science, Guangzhou University, Guangzhou 510006, P. R. China
| | - Xiu-Shuang Yang
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Laboratory of Optoelectronic Materials and Sensor Components, Guangzhou Key Laboratory of Sensing Materials & Devices, Centre for Advanced Analytical Science, Guangzhou University, Guangzhou 510006, P. R. China
| | - Xin Tian
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Laboratory of Optoelectronic Materials and Sensor Components, Guangzhou Key Laboratory of Sensing Materials & Devices, Centre for Advanced Analytical Science, Guangzhou University, Guangzhou 510006, P. R. China
| | - Hao-Yu Ma
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Laboratory of Optoelectronic Materials and Sensor Components, Guangzhou Key Laboratory of Sensing Materials & Devices, Centre for Advanced Analytical Science, Guangzhou University, Guangzhou 510006, P. R. China
| | - Wei Wang
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Laboratory of Optoelectronic Materials and Sensor Components, Guangzhou Key Laboratory of Sensing Materials & Devices, Centre for Advanced Analytical Science, Guangzhou University, Guangzhou 510006, P. R. China
| | - Wen-Sheng Zhang
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Laboratory of Optoelectronic Materials and Sensor Components, Guangzhou Key Laboratory of Sensing Materials & Devices, Centre for Advanced Analytical Science, Guangzhou University, Guangzhou 510006, P. R. China
| | - Dong-Xue Han
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Laboratory of Optoelectronic Materials and Sensor Components, Guangzhou Key Laboratory of Sensing Materials & Devices, Centre for Advanced Analytical Science, Guangzhou University, Guangzhou 510006, P. R. China
| | - Dong-Dong Qin
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Laboratory of Optoelectronic Materials and Sensor Components, Guangzhou Key Laboratory of Sensing Materials & Devices, Centre for Advanced Analytical Science, Guangzhou University, Guangzhou 510006, P. R. China
| | - Dong-Fang Han
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Li Niu
- School of Chemistry and Chemical Engineering, Guangdong Provincial Key Laboratory of Optoelectronic Materials and Sensor Components, Guangzhou Key Laboratory of Sensing Materials & Devices, Centre for Advanced Analytical Science, Guangzhou University, Guangzhou 510006, P. R. China
- School of Chemical Engineering and Technology, Sun Yat-sen University, Zhuhai 519082, P. R. China
| |
Collapse
|
330
|
Xu Y, Xu L, Chen Q, Zou C, Huang J, Zhang L. Crosstalk between exosomes and tumor-associated macrophages in hepatocellular carcinoma: implication for cancer progression and therapy. Front Immunol 2025; 16:1512480. [PMID: 40264760 PMCID: PMC12011854 DOI: 10.3389/fimmu.2025.1512480] [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: 10/16/2024] [Accepted: 02/26/2025] [Indexed: 04/24/2025] Open
Abstract
Hepatocellular carcinoma (HCC), the most prevalent type of primary liver cancer, represents a significant cause of cancer-related mortality. While our understanding of its pathogenesis is comparatively comprehensive, the influence of the tumor microenvironment (TME) on its progression warrants additional investigation. Tumor-associated macrophages (TAMs) have significant impacts on cancer cell proliferation, migration, invasion, and immune response, facilitating a complex interaction within the TME. Exosomes, which measure between 30 and 150 nanometers in size, are categorized into small extracellular vesicles, secreted by a wide range of eukaryotic cells. They can transfer biological molecules including proteins, non-coding RNAs, and lipids, which mediates the intercellular communication within the TME. Emerging evidence has revealed that exosomes regulate macrophage polarization, thus impacting cancer progression and immune responses within the TME of HCC. Moreover, TAM-derived exosomes also play crucial roles in malignant transformation, which hold immense potential for cancer therapy. In this review, we elaborate on the crosstalk between exosomes and TAMs within TME during HCC development. Moreover, we delve into the feasible treatment approaches for exosomes in cancer therapy and emphasize the limitations and challenges for the translation of exosomes derived from TAMs into clinical courses for cancer therapy, which may provide new perspectives on further ameliorations of therapeutic regimes based on exosomes to advance their clinical applications.
Collapse
Affiliation(s)
- Ying Xu
- Department of Anesthesiology Operating Room, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Linyue Xu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Qiuyan Chen
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Can Zou
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Ju Huang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Limei Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
331
|
Hu W, Long Y, Liang W, Zheng H. Integrating Ferroelectric Fields with Active Sites for the Construction of Highly Efficient Nanozymes. Anal Chem 2025; 97:7501-7509. [PMID: 40145867 DOI: 10.1021/acs.analchem.5c00657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Enhancing nanozymes' catalytic activity is challenging yet crucial for practical applications. Herein, inspired by the electrostatic preorganization effect in the catalytic process of natural protein enzymes, a nanozyme is constructed by decorating ferroelectric BaTiO3 nanoparticles (BTO) with hemin, which is often regarded as the active site of natural horseradish peroxidase (HRP). The Hemin-BTO nanozyme demonstrates excellent peroxidase-like (POD-like) activity with the catalytic constant (Kcat) up to 9.71 × 105 s-1 and 1.41 × 106 s-1 for TMB and H2O2 substrates, which is ca. 240-fold and 400-fold greater than that of HRP. Theoretical studies utilizing Density Functional Theory calculations revealed the underlying mechanism. The spontaneous polarization electric field of BTO adjusts the internal electrostatic field of the active site hemin, thereby enhancing the affinity between the Hemin-BTO nanozyme and the substrate. Simultaneously, the existence of hemin reduced the recombination of BTO charge carriers, accelerated electron transfer, and thus promoted the generation of reactive oxygen species, effectively enhancing its POD-like activity. In addition, Hemin-BTO has been successfully used to establish an immunoassay of human brain natriuretic peptide. This work presented a feasible strategy to construct nanozymes with highly catalytic activity by integrating the ferroelectric fields with the active site of natural enzymes.
Collapse
Affiliation(s)
- Weiling Hu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, P. R. China
| | - Yijuan Long
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, P. R. China
| | - Wenbin Liang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, P. R. China
| | - Huzhi Zheng
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, P. R. China
| |
Collapse
|
332
|
Liu Q, Tang R, Chen X, Chen J, Huang Y, Wang S, Gan N, Huang S. Cryogenically Induced Highly Ordered Single-Strand DNA-Modified Magnetic Nanoprobes for Rapid and Ultrasensitive Bioanalysis. Anal Chem 2025; 97:7260-7270. [PMID: 40159104 DOI: 10.1021/acs.analchem.4c06869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The development of highly sensitive detection methods for bioanalysis is crucial for early disease diagnosis. Electrochemical biosensing technology offers unique advantages in this area due to its rapid response, high sensitivity, and low cost. However, achieving efficient and rapid transfer of signaling molecules to the electrode interface to facilitate effective interaction between signal molecules and the sensing surface remains a critical challenge for ultrasensitive electrochemical detection. In this study, we discovered that single-stranded DNA-modified magnetic nanoprobes (signal probe A) subjected to cryogenic treatment can rapidly form an orderly monolayer at the electrode interface under an external magnetic field, while this phenomenon was not observed with double-stranded DNA-modified magnetic nanoprobes (signal probe B). Building on this finding, we developed a signal probe with a protective complementary strand (signal probe B) that, upon interaction with target molecules, is converted into signal probe A. This transformation, combined with cryogenic treatment, enables the ultrasensitive detection of target molecules. Using miRNA-21 and a carcinoembryonic antigen (CEA) as model targets, we optimized the detection conditions, achieving a detection limit as low as 3.4 aM for miRNA-21 and 0.28 fg/mL for CEA with excellent versatility. In summary, this study introduces a highly efficient, rapid, enzyme-free, and environmentally friendly electrochemical signal amplification strategy. This approach not only provides an innovative solution for the ultrasensitive bioanalysis but also offers new insights into enhancing signal molecule-sensor interface interactions in electrochemical biosensors.
Collapse
Affiliation(s)
- Qiwen Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Rentao Tang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiyu Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jiamei Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yang Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Sheng Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Ning Gan
- College of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Shengfeng Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
333
|
Lin S, Lin T, Xu H, Li H, Zhang W, Wu C, Lu S, Chen Y, Han X. Nanoparticles-based electrochemical sensing platform for high-through immunoassay with redox-activity CaCO 3 nanotags on a magnetic microfluidic device. Mikrochim Acta 2025; 192:283. [PMID: 40198434 DOI: 10.1007/s00604-025-07149-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
A new nanoparticles-based sensing platform was designed for high-through electrochemical immunoassay of ferritin (FET) biomarker on a magneto-controlled microfluidic device by using anti-FET capture antibody-conjugated magnetic sensing probes. Thionine-doped calcium carbonate (CaCO3) nanoparticles labeled with anti-FET detection antibodies were utilized as the recognition elements. Introduction of target FET caused the sandwich-type immunoreaction between two antibodies. The formed immunocomplexes were attached onto magnetic microfluidic sensing interface through an external magnet. Subsequently, the carried CaCO3 nanoparticles were dissolved under acidic conditions to release the doped thionine molecules with redox activity. The thionine-based voltammetric signals increased with the increment of target FET levels within the linear range 0.01-100 ng mL-1. The limit of detection was 7.9 pg mL-1 FET. Good analytical properties such as selectivity, reproducibility, and accuracy were achieved with the nanoparticles-based magnetic electrochemical immunoassay. More significantly, the magnetic microfluidic electrochemical immunoassay provides new opportunities for rapid, simple, and cost-effective serum sample analysis.
Collapse
Affiliation(s)
- Shaofeng Lin
- Department of Thoracic Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Ting Lin
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, People's Republic of China
| | - Haipeng Xu
- Department of Thoracic Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Haining Li
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, People's Republic of China
| | - Wenwen Zhang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Chenyu Wu
- The School of Basic Medical Sciences, Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Shuyi Lu
- The School of Basic Medical Sciences, Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Yanping Chen
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China.
| | - Xiao Han
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, People's Republic of China.
| |
Collapse
|
334
|
Peng S, Li C, Wang Y, Yi Y, Chen X, Yin Y, Yang F, Chen F, Ouyang Y, Xu H, Chen B, Shi H, Li Q, Zhao Y, Feng L, Gan Z, Xie X. The metabolic enzyme GYS1 condenses with NONO/p54 nrb in the nucleus and spatiotemporally regulates glycogenesis and myogenic differentiation. Cell Death Differ 2025:10.1038/s41418-025-01509-4. [PMID: 40200092 DOI: 10.1038/s41418-025-01509-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 03/05/2025] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Accumulating evidence indicates that metabolic enzymes can directly couple metabolic signals to transcriptional adaptation and cell differentiation. Glycogen synthase 1 (GYS1), the key metabolic enzyme for glycogenesis, is a nucleocytoplasmic shuttling protein compartmentalized in the cytosol and nucleus. However, the spatiotemporal regulation and biological function of nuclear GYS1 (nGYS1) microcompartments remain unclear. Here, we show that GYS1 dynamically reorganizes into nuclear condensates under conditions of glycogen depletion or transcription inhibition. nGYS1 complexes with the transcription factor NONO/p54nrb and undergoes liquid-liquid phase separation to form biomolecular condensates, leading to its nuclear retention and inhibition of glycogen biosynthesis. Compared to their wild-type littermates, Nono-deficient mice exhibit exercise intolerance, higher muscle glycogen content, and smaller myofibers. Additionally, Gys1 or Nono deficiency prevents C2C12 differentiation and cardiotoxin-induced muscle regeneration in mice. Mechanistically, nGYS1 and NONO co-condense with the myogenic transcription factor MyoD and preinitiation complex (PIC) proteins to form transcriptional condensates, driving myogenic gene expression during myoblast differentiation. These results reveal the spatiotemporal regulation and subcellular function of nuclear GYS1 condensates in glycogenesis and myogenesis, providing mechanistic insights into glycogenoses and muscular dystrophy.
Collapse
Affiliation(s)
- Shujun Peng
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China
| | - Canrong Li
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China
| | - Yifan Wang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China
| | - Yuguo Yi
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China
| | - Xinyu Chen
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China
| | - Yujing Yin
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Medical School of Nanjing University, Nanjing University, Nanjing, PR China
| | - Fan Yang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China
| | - Fengzhi Chen
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China
| | - Yingyi Ouyang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China
| | - Haolun Xu
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China
| | - Baicheng Chen
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China
| | - Haowen Shi
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China
| | - Qingrun Li
- CAS Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, PR China
| | - Yu Zhao
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China
| | - Lin Feng
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| | - Zhenji Gan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Medical School of Nanjing University, Nanjing University, Nanjing, PR China.
| | - Xiaoduo Xie
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, PR China.
| |
Collapse
|
335
|
Zhao Y, Wang X, Yang X, Li J, Han B. Insights into the history and trends of nanotechnology for the treatment of hepatocellular carcinoma: a bibliometric-based visual analysis. Discov Oncol 2025; 16:484. [PMID: 40192866 PMCID: PMC11977073 DOI: 10.1007/s12672-025-02145-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/13/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Nanotechnology has great potential and advantages in the treatment of hepatocellular carcinoma (HCC), but the research trends and future directions are not yet clear. OBJECTIVES Analyze the development trajectory, research hotspots, and future trends of nanotechnology and HCC research globally in the past 20 years, providing a more comprehensive and intuitive reference for researchers in this field. METHODS Retrieve relevant literature on nanotechnology and HCC research in the Web of Science (WOS) Core Collection database, and conduct bibliometric analysis using software such as CiteSpace, VOSviewer, and SCImago Graphica. RESULTS A total of 852 English publications meeting the criteria were retrieved from the WOS database, with an overall increasing trend in the number of publications and citation frequency over the years. China leads in the number of publications and international collaborations, followed by the USA and India. The most influential research institution is the Chinese Academy of Sciences, the most influential scholar/team is the Rahman, Mahfoozur team, and the journal with the most publications is the International Journal of Nanomedicine. A comprehensive analysis reveals that the current main research directions include new types of nanoparticles, targeted drug delivery systems, photothermal/photodynamic therapy, gene delivery systems, diagnostics, and imaging. It is anticipated that further collaboration among scholars, institutions, and countries will accelerate the development of nanotechnology in the field of HCC research. CONCLUSION This study provides an in-depth analysis of the research status and development trends of nanotechnology in treating HCC from a bibliometric perspective, offering possible guidance for researchers to explore hot topics and frontiers, select suitable journals, and partners in this field.
Collapse
Affiliation(s)
- Yulei Zhao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Xingxin Wang
- College of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiaoman Yang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Jiaheng Li
- College of Health, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Bingbing Han
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
| |
Collapse
|
336
|
Piergentili R, Sechi S. Targeting Regulatory Noncoding RNAs in Human Cancer: The State of the Art in Clinical Trials. Pharmaceutics 2025; 17:471. [PMID: 40284466 PMCID: PMC12030637 DOI: 10.3390/pharmaceutics17040471] [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: 01/22/2025] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025] Open
Abstract
Noncoding RNAs (ncRNAs) are a heterogeneous group of RNA molecules whose classification is mainly based on arbitrary criteria such as the molecule length, secondary structures, and cellular functions. A large fraction of these ncRNAs play a regulatory role regarding messenger RNAs (mRNAs) or other ncRNAs, creating an intracellular network of cross-interactions that allow the fine and complex regulation of gene expression. Altering the balance between these interactions may be sufficient to cause a transition from health to disease and vice versa. This leads to the possibility of intervening in these mechanisms to re-establish health in patients. The regulatory role of ncRNAs is associated with all cancer hallmarks, such as proliferation, apoptosis, invasion, metastasis, and genomic instability. Based on the function performed in carcinogenesis, ncRNAs may behave either as oncogenes or tumor suppressors. However, this distinction is not rigid; some ncRNAs can fall into both classes depending on the tissue considered or the target molecule. Furthermore, some of them are also involved in regulating the response to traditional cancer-therapeutic approaches. In general, the regulation of molecular mechanisms by ncRNAs is very complex and still largely unclear, but it has enormous potential both for the development of new therapies, especially in cases where traditional methods fail, and for their use as novel and more efficient biomarkers. Overall, this review will provide a brief overview of ncRNAs in human cancer biology, with a specific focus on describing the most recent ongoing clinical trials (CT) in which ncRNAs have been tested for their potential as therapeutic agents or evaluated as biomarkers.
Collapse
|
337
|
Regeni I, Bonnet S. Supramolecular approaches for the treatment of hypoxic regions in tumours. Nat Rev Chem 2025:10.1038/s41570-025-00705-7. [PMID: 40185999 DOI: 10.1038/s41570-025-00705-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 04/07/2025]
Abstract
Supramolecular chemistry provides a range of 'weak' intermolecular interactions that allow drugs and prodrugs to self-assemble. In the complex biological setting of blood and tumours, these interactions must be stable enough for efficient and selective drug delivery to the tumour site, but weak enough to allow the release of the cytotoxic load. The non-covalent nature of supramolecular interactions enables the detachment of smaller (pro)drug monomers that can penetrate cancer cells differently to the original nanoparticles. Hypoxic tumours show low oxygen levels due to poor vascularization, which poses challenges for drug delivery and generates biological resistances. Supramolecular building blocks specifically designed for hypoxic tumours offer targeted activation of prodrug self-assemblies, enhancing effectiveness against hypoxic cancer cells and hypoxic regions in tumours. This Review explores how supramolecular chemistry can improve (pro)drug delivery and activation in hypoxic tumours.
Collapse
Affiliation(s)
- Irene Regeni
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.
| | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
338
|
Zhao X, Hou S, Hao R, Zang Y, Song D. Prognostic significance of circulating tumor DNA detection and quantification in cervical cancer: a systematic review and meta-analysis. Front Oncol 2025; 15:1566750. [PMID: 40255423 PMCID: PMC12006000 DOI: 10.3389/fonc.2025.1566750] [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: 01/25/2025] [Accepted: 03/19/2025] [Indexed: 04/22/2025] Open
Abstract
Background Circulating tumor DNA (ctDNA) is an emerging biomarker in cervical cancer, with elevated levels typically indicating a higher tumor burden. However, its prognostic value in cervical cancer patients remains debated. This meta-analysis aims to clarify the prognostic significance of ctDNA in this patient population. Methods We searched the PubMed, Cochrane Library, CNKI, and EMBASE databases for studies published up to September 30, 2024, to investigate the prognostic significance of ctDNA in cervical cancer patients. The outcome measures included overall survival (OS) and progression-free survival (PFS)/disease-free survival (DFS). Results This analysis included 10 studies encompassing a total of 706 cervical cancer patients. Findings revealed that patients with detectable baseline ctDNA had significantly poorer OS(HR = 1.64, 95% CI = 1.45-1.86, P < 0.001) as well as worse PFS or DFS (HR = 1.42, 95% CI = 1.07-1.89, P = 0.015). Additionally, ctDNA detectability during treatment was strongly associated with poorer OS (HR = 17.22, 95% CI = 4.43-66.89, P < 0.001) and PFS/DFS (HR = 4.16, 95% CI = 2.57-6.73, P < 0.001). Conclusions This meta-analysis demonstrates that elevated ctDNA levels are significantly associated with poorer PFS, DFS, and OS in patients with cervical cancer. However, data regarding the association between ctDNA levels and OS are relatively limited, and the number of included studies remains small, with a potential risk of publication bias. Based on the current evidence, ctDNA shows promise as a valuable tool for pre-treatment assessment and an effective biomarker for monitoring therapeutic response and disease progression. Further large-scale, prospective studies are warranted to validate these findings and establish their reliability and clinical applicability. Systematic Review Registration inplasy.com, identifier INPLASY2024120083.
Collapse
Affiliation(s)
- Xiumin Zhao
- Department of Neurology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shufu Hou
- Department of Gastrointestinal Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ruiqi Hao
- Department of Gastrointestinal Surgery, Xintai City People’s Hospital, Xintai, Shandong, China
| | - Yelei Zang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Dandan Song
- Department of Neurology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
339
|
Liu H, Chen L, Chen Y, Jin Y, Chen X, Ma N, Yang F, Bi H, Wen X, Xu S, Chen J, Lin Y, Yang Y, Wu Y, Chen Y. TCP1 promotes the progression of malignant tumours by stabilizing c-Myc through the AKT/GSK-3β and ERK signalling pathways. Commun Biol 2025; 8:563. [PMID: 40185866 PMCID: PMC11971430 DOI: 10.1038/s42003-025-07867-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/03/2025] [Indexed: 04/07/2025] Open
Abstract
The chaperonin tailless complex polypeptide 1 (TCP1) is a key subunit of chaperonin containing TCP1 (CCT) that regulates the folding and stability of proteins during cancer progression. Here, the prognostic significance of TCP1 was explored mainly in patients with hepatocellular carcinoma (HCC) and pancreatic ductal adenocarcinoma (PDAC). We showed that TCP1 expression was significantly greater in clinically malignant tumour tissues than in normal tissues and that high TCP1 expression was associated with poor prognosis. TCP1 suppression not only decreased the proliferation and invasion of cancer cells in vitro but also inhibited tumour growth and metastasis in vivo. The underlying mechanisms were determined by ubiquitination assays and Co-IP (Co-Immunoprecipitation) experiments, and it was found that TCP1 regulated the stability of c-Myc through the RAC-alpha serine/threonine-protein kinase (AKT) /Glycogen synthase kinase 3β (GSK-3β) and extracellular regulated protein kinases (ERK) signalling pathways. Moreover, TCP1 knock-in (TCP1-KI) dramatically promoted the occurrence of diethylnitrosamine (DEN) -induced primary HCC in mice. Our results highlight the critical role of TCP1 in HCC and PDAC and reveal a novel mechanism to suppress HCC and PDAC by targeting c-Myc via the TCP1-induced promotion of the AKT/GSK-3β and ERK signalling pathways. TCP1 is able to modulate the stability of target proteins by multiple pathways, thus promoting the progression of HCC and PDAC. Our study identifies TCP1 as a prognostic novel marker and therapeutic target of HCC and PDAC.
Collapse
Affiliation(s)
- Hekun Liu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Linying Chen
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, No. 20, Chazhong Road, 350005, Fuzhou, Fujian, China
| | - Yuwen Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Yiyi Jin
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Xiance Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Nengjun Ma
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Fan Yang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Huixia Bi
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Xinxin Wen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Shenmin Xu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Juan Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Yanping Lin
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Yinghong Yang
- Department of Pathology, Fujian Medical University Union Hospital, No. 29, Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Yong Wu
- Fujian Institute of Haematology, Fujian Key Laboratory on Haematology, Fujian Medical University Union Hospital, No. 29, Xinquan Road, 350001, Fuzhou, Fujian, China.
| | - Yuanzhong Chen
- Fujian Institute of Haematology, Fujian Key Laboratory on Haematology, Fujian Medical University Union Hospital, No. 29, Xinquan Road, 350001, Fuzhou, Fujian, China.
| |
Collapse
|
340
|
Jin Z. Comment on "Peripheral T Lymphocyte Predicts the Prognosis of Gastric Cancer Patients Undergoing Radical Gastrectomy: a Multicenter Retrospective Cohort Study" [Letter]. J Inflamm Res 2025; 18:4613-4614. [PMID: 40195956 PMCID: PMC11974999 DOI: 10.2147/jir.s523648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025] Open
Affiliation(s)
- Zhihui Jin
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital, College of Medicine, Zhejiang University, Ningbo, People’s Republic of China
- Department of Hematology and Oncology, Beilun People’s Hospital, Ningbo, People’s Republic of China
| |
Collapse
|
341
|
Pu H, Huang J, Gui B, Chen Y, Guo Y, Lian Y, Pan J, Hu Y, Jiang N, Deng Q, Zhou Q. Ultrasound-Responsive Nanobubbles for Breast Cancer: Synergistic Sonodynamic, Chemotherapy, and Immune Activation through the cGAS-STING Pathway. ACS APPLIED MATERIALS & INTERFACES 2025; 17:19317-19334. [PMID: 40126217 DOI: 10.1021/acsami.4c21493] [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: 03/25/2025]
Abstract
Breast cancer remains the leading cause of cancer-related deaths among women worldwide, necessitating more effective treatment strategies. Chemotherapy combined with immunotherapy is the first-line treatment for breast cancer, but it still suffers from limited therapeutic efficiency and serious side effects, which are usually due to the poor delivery efficiency, drug resistance of tumor cells, and immunosuppressive tumor microenvironment. This study explores the development of ultrasound-responsive nanobubbles (Ce6/PTX Nbs) for targeted imaging and sonoimmunotherapy in breast cancer treatment. By integrating sonodynamic therapy (SDT), chemotherapy, and immunotherapy, the nanobubbles aim to address challenges such as poor drug delivery, systemic toxicity, and immune suppression in conventional therapies. The nanobubbles, composed of sonosensitizer chlorin e6 (Ce6)-modified phospholipid and loaded with the chemotherapeutic agent paclitaxel (PTX) enhancing drug-loading capacity, are designed to precisely target tumor sites via cyclic-RGD peptides. Upon ultrasound activation, Ce6 induces reactive oxygen species (ROS), promoting immunogenic cell death (ICD), while PTX disrupts tumor cell mitosis, enhancing the immune response. The nanobubbles' ultrasound responsiveness facilitates real-time imaging and controlled drug release, maximizing therapeutic efficacy while minimizing side effects. Key findings demonstrate that Ce6/PTX Nbs significantly reduced tumor growth in a 4T1 breast cancer model, enhanced immune activation via the cGAS-STING pathway, and increased the infiltration of CD8+ T cells in both primary and distant tumors. In combination with anti-PD-L1 checkpoint inhibitors, the treatment achieved a substantial suppression of tumor metastasis. This innovative approach offers a highly targeted, effective, and minimally toxic breast cancer treatment with potential for clinical translation due to its dual imaging and therapeutic capabilities.
Collapse
Affiliation(s)
- Huan Pu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Jia Huang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Bin Gui
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Yueying Chen
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Yuxin Guo
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Yingtao Lian
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Juhong Pan
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Yugang Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Nan Jiang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Qing Deng
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Qing Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| |
Collapse
|
342
|
Wang X, Li J, Luo Z, Gao Z, Huang Y, Luo J, Wang X, Zhang Y, Tan M, Hou Z. ATP-Exhausted Strategy Induced Anti-Tumor Low-Temperature Photothermal Therapy Based on Rare Earth Nanocrystals Modified Hollow Porous MnO x Nanozyme with TME-Activated NIR-II Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410070. [PMID: 40025926 DOI: 10.1002/smll.202410070] [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: 10/28/2024] [Revised: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Insufficient adenosine triphosphate (ATP) can reduce the synthesis of heat-stress-induced heat shock proteins (HSPs) to promote the efficiency of mild photothermal therapy (mPTT), thus the rational design of ATP-exhausted strategy based on nanotechnology is an effective approach to resuscitate mPTT. Herein, Nd3+ doped nanocrystals (NaYF4:Nd@CaF2, Nd-NCs) modified hollow mesoporous manganese oxide (H-MnOx) nanocomposite (H-MnOx@Nd-NCs, MN) is synthesized, and loaded with glucose transporters (GLUTs) inhibitor KL-11743, noted as MN-KL nanozyme. In tumor microenvironment (TME), MN-KL can react with overexpressed glutathione (GSH) to release KL-11743, which can suppress the synthesis of intracellular ATP at the source by blocking glucose uptake to inhibit HSPs expression, meanwhile, MN-KL catalyzes the production of ·O2 -/1O2/·OH and lipid peroxidation (LPO) to cleave existing HSPs. Through a two-pronged strategy with ATP inhibition and oxide accumulation, reducing the level of HSPs can be guaranteed for achieving efficient mPTT in both subcutaneous and in situ tumor models in mice. During this process, Nd-NCs can absorb near-infrared light and convert it into heat, and the quenched fluorescence of Nd-NCs by H-MnOx can be recovered through GSH-triggered biodegradation in tumors, thus the modification of Nd-NCs not only provides photothermal effect but also enables MN to own TME-activated fluorescence imaging.
Collapse
Affiliation(s)
- Xiaozhao Wang
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, P. R. China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, P. R. China
| | - Jing Li
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, P. R. China
| | - Zhengtao Luo
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Zhimin Gao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Yongxin Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Jiamin Luo
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Xinyi Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Yaru Zhang
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, P. R. China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, 511518, P. R. China
| | - Meiling Tan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Zhiyao Hou
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, P. R. China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, 511518, P. R. China
| |
Collapse
|
343
|
Mohammed MA, Hay NHA, Mohammed MT, Mahmoud HS, Ahmed MY, Abdelmenem A, Abdelrahim DS. The effect of adipose-derived mesenchymal stem cells against high fructose diet induced liver dysfunction and dysbiosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4525-4537. [PMID: 39500806 PMCID: PMC11978704 DOI: 10.1007/s00210-024-03518-5] [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: 06/03/2024] [Accepted: 10/05/2024] [Indexed: 04/10/2025]
Abstract
High fructose diet (HFrD) has been approved to be involved in the pathogenesis of insulin resistance. Mesenchymal stem cells have a vital role in the treatment of various diseases including metabolic disturbances. We investigated the effect of Adipose-derived mesenchymal stem cells (ADMSCs) against HFrD-induced metabolic disorders and the molecular mechanisms for this effect. Rats were divided into 3 groups; control, HFrD, and combined HFrD with ADMSCs. We assessed liver functions, gut microbiota activity, oxidative stress, adiponectin, and IL10 levels. Also, we measured SREBP-1, IRS-1 expression using Western blot, and Malat1 expression using rt-PCR. ADMSCs antagonized metabolic abnormalities induced by HFrD in the form of improvement of liver functions and alleviation of oxidative stress. In addition, ADMSCs ameliorated gut microbiota activity besides the elevation of adiponectin and IL10 levels. ADMSCs attenuated insulin resistance through upregulation of IRS1 and downregulation of SREBP-1 and Malat1. ADMSCs can protect against HFrD-induced metabolic hazards.
Collapse
Affiliation(s)
| | - Nesma Hussein Abel Hay
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Maha Tarek Mohammed
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hoda Sayed Mahmoud
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Manar Yehia Ahmed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed Abdelmenem
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Dina Sayed Abdelrahim
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Department of Pharmacology, Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
| |
Collapse
|
344
|
Yan C, Zhu W, Li R, Xu Q, Li D, Zhang W, Leng L, Shao A, Guo Z. Mapping Dynamic Protein Clustering with AIEgen-Active Chemigenetic Probe. Angew Chem Int Ed Engl 2025; 64:e202422996. [PMID: 39831846 DOI: 10.1002/anie.202422996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/13/2025] [Accepted: 01/18/2025] [Indexed: 01/22/2025]
Abstract
Protein clustering/disassembling is a fundamental process in biomolecular condensates, playing a crucial role in cell fate decision and cellular homeostasis. However, the inherent features of protein clustering, especially for its reversible behavior and subtle microenvironment variation, present significant hurdles in probe chemistry for tracking protein clustering dynamics. Herein, we report a bilateral-tailored chemigenetic probe, in which an "amphiphilic" aggregate-induced emission luminogen (AIEgen) QMSO3Cl is covalently conjugated to a protein tag that is genetically fused to protein-of-interest (POI). Prior to target POI, the "amphiphilic" AIE-active QMSO3Cl achieves a completely dark state in both aqueous biological environment and lipophilic organelles, thereby ensuring an ultra-low intrinsic background interference. Upon reaching POI, the combination of synthetic molecule and genetically encoded protein allows for protein clustering-dependent ultra-sensitive response, with a substantial lighting-up fluorescence (67.5-fold) as protein transitions from disassembling to clustering state. Such ultra-high signal-to-noise ratio enables to monitor the dynamic and fate of inositol requiring enzyme 1 (IRE1) clustering/disassembling under both acute and chronic endoplasmic reticulum (ER) stress in living cells. For the first time, we have demonstrated the use of chemigenetic probe to reveal therapy-induced ER stress and screen drugs in a three-dimensional scenario: microviscosity change, clustering dynamic, and cluster morphology. This chemigenetic probe design strategy would greatly facilitate the advancement of mapping protein dynamics in cell homeostasis and medicine research.
Collapse
Affiliation(s)
- Chenxu Yan
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Wendi Zhu
- Stem Cell and Regenerative Medicine Lab, Institute of Clinical Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Runqi Li
- Key Laboratory of Carbohydrate Vaccines and Drugs in Jiangsu Province, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Qin Xu
- Key Laboratory of Carbohydrate Vaccines and Drugs in Jiangsu Province, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Dan Li
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Weixu Zhang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Ling Leng
- Stem Cell and Regenerative Medicine Lab, Institute of Clinical Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Andong Shao
- Key Laboratory of Carbohydrate Vaccines and Drugs in Jiangsu Province, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Zhiqian Guo
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai, 200237, China
| |
Collapse
|
345
|
Wang T, Zhao J, Chen M, Wu D, Wu Z, Wu Y, Chen M, Gan B. On-demand celastrol delivery by hyaluronic acid-porphyrinic metal-organic frameworks for synergistic sonodynamic/pharmacological antibacterial therapy. Int J Biol Macromol 2025; 302:140421. [PMID: 39884622 DOI: 10.1016/j.ijbiomac.2025.140421] [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: 09/04/2024] [Revised: 01/08/2025] [Accepted: 01/26/2025] [Indexed: 02/01/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the most prevalent bacterial pathogens. The multi-drug resistance and strong biofilm-forming ability make the treatment of MRSA infections challenging. It is urgent to develop antibiotic-free, noninvasive and effective strategies against MRSA infections. Recently, celastrol (CLT), a natural phytochemical, showed potent antimicrobial activity against planktonic MRSA. However, its further development was hindered by low solubility, short plasma half-life, poor bioavailability and severe systemic toxicity. To address these issues, hyaluronic acid-porphyrinic metal-organic frameworks loaded with CLT (HA@PCN-CLT) were fabricated for combating MRSA biofilms. Once encountering the overexpressed HAases in biofilm, their surface charge reversed to positive to facilitate biofilm penetration under ultrasound irradiation, which was revealed by 3D fluorescence image. In vitro release profiles suggested that HA@PCNCLT exhibited ultrasound promoted pH/HAase-responsive release properties. Taking advantage of superior sonodynamic performance and on-demand CLT release, HA@PCN-CLT NPs effectively reduced biofilm biomass and killed planktonic and biofilm-embedded MRSA. Moreover, HA@PCN-CLT NPs completely eradicated the MRSA infection in mice under ultrasound irradiation and promoted abscess healing by stimulating angiogenesis and collagen deposition, accompanied by excellent biocompatibility and negligible toxicity in vivo. HA@PCN-CLT NPs provided a promising antibiotic-free strategy against MRSA biofilms through synergistic combination of sonodynamic therapy and phytochemicals.
Collapse
Affiliation(s)
- Tao Wang
- Institute of urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu National Agricultural Science &Technology Center, Chengdu 610213, China
| | - Jin Zhao
- Institute of urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu National Agricultural Science &Technology Center, Chengdu 610213, China
| | - Mengxing Chen
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Dan Wu
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Zhi Wu
- Institute of urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu National Agricultural Science &Technology Center, Chengdu 610213, China
| | - Yiyou Wu
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Maohua Chen
- Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya 572025, China.
| | - Bingcheng Gan
- Institute of urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu National Agricultural Science &Technology Center, Chengdu 610213, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China.
| |
Collapse
|
346
|
Magyar CTJ, Rajendran L, Li Z, Banz V, Vogel A, O'Kane GM, Chan ACY, Sapisochin G. Precision surgery for hepatocellular carcinoma. Lancet Gastroenterol Hepatol 2025; 10:350-368. [PMID: 39993401 DOI: 10.1016/s2468-1253(24)00434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 02/26/2025]
Abstract
Hepatocellular carcinoma arises in the setting of cirrhosis in most cases, requiring multidisciplinary input to define resectability. In this regard, more precise surgical management considers patient factors and anatomical states, including resection margins, tumour biology, and perioperative therapy. Together with advances in surgical techniques, this integrated approach has resulted in considerable improvements in patient morbidity and oncological outcomes. Despite this, recurrence rates in hepatocellular carcinoma remain high. As the systemic treatment landscape in hepatocellular carcinoma continues to evolve and locoregional options are increasingly used, we review current and future opportunities to individualise the surgical management of patients with hepatocellular carcinoma.
Collapse
Affiliation(s)
- Christian Tibor Josef Magyar
- HPB Surgical Oncology, University Health Network, Toronto, ON, Canada; Multi-Organ Transplant Program, University Health Network, Toronto, ON, Canada; Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Luckshi Rajendran
- HPB Surgical Oncology, University Health Network, Toronto, ON, Canada; Multi-Organ Transplant Program, University Health Network, Toronto, ON, Canada; Division of Transplant Surgery, Henry Ford Hospital, Detroit, MI, USA
| | - Zhihao Li
- HPB Surgical Oncology, University Health Network, Toronto, ON, Canada; Multi-Organ Transplant Program, University Health Network, Toronto, ON, Canada
| | - Vanessa Banz
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Arndt Vogel
- Medical Oncology, Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada; Division of Gastroenterology and Hepatology, Toronto General Hospital, Toronto, ON, Canada; Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hanover, Germany
| | - Grainne Mary O'Kane
- Medical Oncology, Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada; Department of Medicine Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; St Vincent's University Hospital and School of Medicine, University College Dublin, Dublin, Ireland
| | - Albert Chi-Yan Chan
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Gonzalo Sapisochin
- HPB Surgical Oncology, University Health Network, Toronto, ON, Canada; Multi-Organ Transplant Program, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
347
|
Zhang R, Li Y, Guan F, Fu G, Liu P, Bai X, Yang Y, Sun C, Zhang T. A homogalacturonan-rich pectic polysaccharide isolated from Lonicera japonica Thunb. modulates galectin-4-mediated bioactivity and anti-hepatocellular carcinoma activity. Int J Biol Macromol 2025; 302:140618. [PMID: 39900157 DOI: 10.1016/j.ijbiomac.2025.140618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/05/2025]
Abstract
L. japonica is a traditional Chinese medicine with dual-use properties. Herein, a HG-rich pectic polysaccharide, WLJP-03A, was purified from the dried flowers of L. japonica, which composed of Rha (5 %), GalA (60 %), Gal (5 %), and Ara (30 %), with a molecular weight of 28.1 kDa. WLJP-03A could be defined as an HG backbone with α-(1 → 3,5)-linked and α-(1 → 5)-linked arabinan, β-(1 → 3,6)-linked and β-(1 → 3)-linked galactan, and type II arabinogalactan side chains. Its interaction with two truncated structural domain proteins of galectin-4 (Gal-4) revealed stronger binding of WLJP-03A to Gal-4C (MIC = 15 μg/mL) than to Gal-4N (MIC = 62 μg/mL), indicating that WLJP-03A mainly interacted with the C-terminal CRD to inhibit the biological activity of Gal-4. Furthermore, in vitro antitumor assays showed that WLJP-03A could inhibit the cellular proliferation and migration of HCCLM3 cells induced by Gal-4. These results provide new insights into the structure-activity relationship between L. japonica polysaccharide and Gal-4.
Collapse
Affiliation(s)
- Renqun Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China; School of Laboratory Medicine, Zunyi Medical University, Zunyi 563006, China
| | - Yiqing Li
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China; Department of Laboratory Medicine, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Fanqi Guan
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China; School of Laboratory Medicine, Zunyi Medical University, Zunyi 563006, China
| | - Guixia Fu
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China; School of Laboratory Medicine, Zunyi Medical University, Zunyi 563006, China
| | - Ping Liu
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563006, China
| | - Xinyu Bai
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563006, China
| | - Yan Yang
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China; School of Laboratory Medicine, Zunyi Medical University, Zunyi 563006, China
| | - Chengxin Sun
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563006, China
| | - Tao Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China; School of Laboratory Medicine, Zunyi Medical University, Zunyi 563006, China.
| |
Collapse
|
348
|
Rozman M, Lukšič M. Electrode Materials for Flexible Electrochromics. Int J Mol Sci 2025; 26:3260. [PMID: 40244101 PMCID: PMC11989306 DOI: 10.3390/ijms26073260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Flexible electrochromic devices (ECDs) represent a distinctive category in optoelectronics, leveraging advanced materials to achieve tunable coloration under applied electric voltage. This review delves into recent advancements in electrode materials for ECDs, with a focus on silver nanowires, metal meshes, conductive polymers, carbon nanotubes, and transparent conductive ceramics. Each material is evaluated based on its manufacturing methods and integration potential. The analysis highlights the prominent role of transparent conductive ceramics and conductive polymers due to their versatility and scalability, while also addressing challenges such as environmental stability and production costs. Use of other alternative materials, such as metal meshes, carbon materials, nanowires and others, are presented here as a comparison as well. Emerging hybrid systems and advanced coating techniques are identified as promising solutions to overcome limitations regarding flexibility and durability. This review underscores the critical importance of electrode innovation in enhancing the performance, sustainability, and application scope of flexible ECDs for next-generation technologies.
Collapse
Affiliation(s)
- Martin Rozman
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, 1000 Ljubljana, Slovenia;
| | | |
Collapse
|
349
|
May JF, Gonske SJ. Insights into mechanisms and significance of domain swapping from emerging examples in the Mog1p/PsbP-like fold. Biochem Biophys Res Commun 2025; 755:151570. [PMID: 40048759 PMCID: PMC11963792 DOI: 10.1016/j.bbrc.2025.151570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/24/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Three-dimensional (3D) domain swapping in proteins occurs when identical polypeptide chains exchange structural elements to form a homo-oligomeric protein. Domain swapping can play a regulatory role for certain oligomeric proteins and has been implicated in deleterious protein aggregation. Here, we examine recently reported 3D domain swapping in proteins that contain the Mog1p/PsbP-like fold, which is a small fold found in non-enzymatic proteins that participate in a variety of distinct cellular processes. This fold was initially identified from structures of the yeast Mog1p protein, which regulates nuclear protein transport in eukaryotes, and PsbP proteins, which are part of photosystem II in plants, green algae, and cyanobacteria. The core structural element of the Mog1p/PsbP-like fold is an α-β-α sandwich that contains a 6- or 7-stranded antiparallel β-sheet. Additionally, most Mog1p/PsbP-like proteins contain an N-terminal β-hairpin that interacts with the α-β-α sandwich. Interestingly, domain-swapped dimers can form by exchange of this N-terminal β-hairpin in certain proteins. We discuss biochemical mechanisms and explore the functional significance of domain-swapping in the formation of an interaction interface in homo-dimers that bind a protein target. Lastly, we examine domain swapping between 2 tandem Mog1p/PsbP-like domains in a multidomain protein. In summary, this review provides recent examples of domain-swapping in proteins containing the Mog1p/PsbP-like fold and highlights general roles for domain-swapping in facilitating protein-protein interactions and in the evolution of multidomain proteins.
Collapse
Affiliation(s)
- John F May
- Department of Chemistry and Biochemistry, University of Wisconsin-La Crosse, 1725 State Street, La Crosse, WI, 54601, USA.
| | - Sara J Gonske
- Department of Chemistry and Biochemistry, University of Wisconsin-La Crosse, 1725 State Street, La Crosse, WI, 54601, USA
| |
Collapse
|
350
|
Liu YC, Feng GL, Jie JL, Zhou W, Liu GJ, Zhang Y, Su HM, Xing GW. Hepatoma Metastasis-Inhibiting Supramolecular Nanoglycocalyx for Enhanced Type I Photodynamic Therapy. Adv Healthc Mater 2025; 14:e2404253. [PMID: 40045640 DOI: 10.1002/adhm.202404253] [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: 10/30/2024] [Revised: 02/25/2025] [Indexed: 04/08/2025]
Abstract
Type I photodynamic therapy (PDT) is well demonstrated to have low oxygen dependency. However, fully suppressing the risk of hypoxia-induced tumor metastasis during PDT remains a great challenge. In this study, a tetra-lactosylated amphiphilic Aza-BODIPY glycocluster (TLBP) is reported that self-assembles into a supramolecular nanoglycocalyx on hepatoma cell membranes, serving as an artificial extracellular matrix (ECM) to inhibit hepatoma metastasis while facilitating efficient Type I PDT. Molecular engineering demonstrates that multi-glycosylation promotes the transition of nanostructures from disordered to ordered self-assembly by regulating intermolecular interactions. This modification enables the TLBP glycocalyx to exhibit significant intermolecular electron transfer, generating superoxide anion radicals (O2 -•) for Type I PDT. Moreover, the TLBP glycocalyx inhibits the PI3K-Akt signaling pathway by reducing Na+/K+-ATPase activity, leading to decreased migration and invasion of HepG2 cells. The synergistic antitumor effect of TLBP glycocalyx is further verified in a HepG2-bearing mouse model. This work innovatively utilizes glycosylation to regulate microelectronic properties and macroscopic nanoscale self-assembly characteristics, providing a novel concept for developing efficient synergistic anti-hepatoma strategies.
Collapse
Affiliation(s)
- Yi-Chen Liu
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Gai-Li Feng
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Jia-Long Jie
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Wei Zhou
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Guang-Jian Liu
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Yuan Zhang
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Hong-Mei Su
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Guo-Wen Xing
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| |
Collapse
|