1
|
Musumeci F, Fasce A, Falesiedi M, Oleari F, Grossi G, Carbone A, Schenone S. Approaching Gallium-68 radiopharmaceuticals for tumor diagnosis: a Medicinal Chemist's perspective. Eur J Med Chem 2025; 294:117760. [PMID: 40393260 DOI: 10.1016/j.ejmech.2025.117760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/22/2025]
Abstract
Nuclear medicine has revolutionized disease diagnosis and treatment, particularly in oncology, by enabling precise imaging and targeted therapies using radiopharmaceuticals. Recently, Gallium-68 (68Ga) has emerged as a powerful positron emission tomography (PET) imaging agent, with a growing role in theranostics when paired with 177Lu for cancer treatment. The ability to obtain 68Ga from 68Ge/68Ga generators, along with its favorable radiochemical and pharmacokinetic properties, has driven an increasing number of clinical applications, which culminated with the approvals of 68Ga-DOTA-TOC and 68Ga-DOTA-TATE for the treatment of neuroendocrine tumors, and 68Ga-PSMA-11 for prostate cancer over the past decade. This review provides a comprehensive overview of 68Ga radiochemistry, chelators, and key compounds in clinical trials, highlighting the potential of this radionuclide in precision oncology.
Collapse
Affiliation(s)
- Francesca Musumeci
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy.
| | - Alessandro Fasce
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy
| | - Marta Falesiedi
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy
| | - Federica Oleari
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy
| | - Giancarlo Grossi
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy
| | - Anna Carbone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy
| | - Silvia Schenone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy
| |
Collapse
|
2
|
Zhang Z, Zhang Q, Wang Y. CAF-mediated tumor vascularization: From mechanistic insights to targeted therapies. Cell Signal 2025; 132:111827. [PMID: 40288665 DOI: 10.1016/j.cellsig.2025.111827] [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: 01/21/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are a major component of the tumor microenvironment (TME) and play a crucial role in tumor progression. The biological properties of tumors, such as drug resistance, vascularization, immunosuppression, and metastasis are closely associated with CAFs. During tumor development, CAFs contribute to tumor progression by remodeling the extracellular matrix (ECM), inhibiting immune cell function, promoting angiogenesis, and facilitating tumor cell growth, invasion, and metastasis. Studies have shown that CAFs can promote endothelial cell proliferation by directly secreting cytokines such as vascular endothelial growth factor (VEGF) and fibroblast Growth Factor (FGF), as well as through exosomes. CAFs also secrete the chemokine stromal cell-derived factor 1 (SDF-1) to recruit endothelial progenitor cells (EPCs) into the peripheral blood and guide their migration to the tumor periphery. Additionally, CAFs can induce tumor cells to transform into "endothelial cells" that participate in vascular wall formation. However, the precise mechanisms remain to be further investigated. Due to their widespread presence in various solid tumors and their tumor-promoting function, CAFs are emerging as therapeutic targets. In this review, we summarize the specific mechanisms through which CAFs promote angiogenesis and outline current therapeutic strategies targeting CAF-induced vascularization, ongoing clinical trials targeting CAFs, and discuss potential future treatment approaches. We hope this will contribute to the advancement of CAF-targeted tumor treatment strategies.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Qing Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yang Wang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
3
|
Ghosh D, Guin A, Kumar A, Das A, Paul S. Comprehensive insights of etiological drivers of hepatocellular carcinoma: Fostering targeted nano delivery to anti-cancer regimes. Biochim Biophys Acta Rev Cancer 2025; 1880:189318. [PMID: 40222420 DOI: 10.1016/j.bbcan.2025.189318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
Hepatocellular carcinoma (HCC) stands as one of the most prevalent and deadliest malignancies on a global scale. Its complex pathogenesis arises from multifactorial etiologies, including viral infections, metabolic syndromes, and environmental carcinogens, all of which drive genetic and molecular aberrations in hepatocytes. This intricate condition is associated with multiple causative factors, resulting in the abnormal activation of various cellular and molecular pathways. Given that HCC frequently manifests within the context of a compromised or cirrhotic liver, coupled with the tendency of late-stage diagnoses, the overall prognosis tends to be unfavorable. Systemic therapy, especially conventional cytotoxic drugs, generally proves ineffective. Despite advancements in therapeutic interventions, conventional treatments such as chemotherapy often exhibit limited efficacy and substantial systemic toxicity. In this context, nanomedicine, particularly lipid-based nanoparticles (LNPs), has emerged as a promising strategy for enhancing drug delivery specificity and reducing adverse effects. This review provides a comprehensive overview of the molecular and metabolic underpinnings of HCC. Furthermore, we explored the role of lipid-based nano-formulations including liposomes, solid lipid nanoparticles, and nanostructured lipid carriers in targeted drug delivery for HCC. We have highlighted recent advances in LNP-based delivery approaches, FDA-approved drugs, and surface modification strategies to improve liver-specific delivery and therapeutic efficacy. It will provide a comprehensive summary of various treatment strategies, recent clinical advances, receptor-targeting strategies and the role of lipid composition in cellular uptake. The review concludes with a critical assessment of existing challenges and future prospects in nanomedicines-driven HCC therapy.
Collapse
Affiliation(s)
- Dipanjan Ghosh
- Department of Biotechnology and Dr. B.C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata 700019, West Bengal, India
| | - Aharna Guin
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India
| | - Aryan Kumar
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India
| | - Amlan Das
- Department of Microbiology & Department of Biochemistry, Royal School of Biosciences, The Assam Royal Global University, Guwahati 781035, Assam, India.
| | - Santanu Paul
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India.
| |
Collapse
|
4
|
Palanisamy B, Mandal AKA. Unlocking the potential: Receptor-mediated targeted drug delivery in cancer therapy. Pathol Res Pract 2025; 270:155955. [PMID: 40209568 DOI: 10.1016/j.prp.2025.155955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/29/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Receptor-mediated targeted drug delivery has emerged as a pivotal strategy in cancer therapy, offering precision and specificity in combating malignant diseases while minimizing systemic toxicity. This review explores the multifaceted role of receptors in cancer biology, emphasizing their contributions to cancer progression, metastasis, and their potential as therapeutic targets. Ligand-based targeting approaches highlight the utility of small molecules, peptides, and antibodies, as well as the development of novel targeting ligands. A critical focus is placed on engineering receptor-targeted nanoparticles and advanced drug delivery systems. Innovations in dual-targeting strategies and the targeted delivery to the tumour microenvironment (TME) and metastatic niches are discussed, underscoring their potential to enhance therapeutic efficacy. Additionally, receptor-targeted imaging is reviewed for its dual role in diagnosis and real-time treatment monitoring. To address the challenges of side effects and off-target toxicity, strategies that minimize these risks while targeting overexpressed receptors in solid tumours are explored. Finally, the review outlines future directions in receptor-targeted cancer therapy, emphasizing the need for interdisciplinary research to refine these strategies further. This comprehensive analysis aims to provide a roadmap for advancing receptor-based therapeutic approaches, ultimately improving outcomes for cancer patients.
Collapse
Affiliation(s)
- Balaji Palanisamy
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| | - Abul Kalam Azad Mandal
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
5
|
Chen WS, Concio CAP, Chang TT, Chen CL, Perez SJLP, Li WS. Optimizing ST6GAL1 inhibition and selectivity using lithocholic acid-amino acid conjugates for antimetastatic and antiangiogenic agent development. Bioorg Chem 2025; 159:108401. [PMID: 40174529 DOI: 10.1016/j.bioorg.2025.108401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/04/2025]
Abstract
A series of LCA-aromatic amino acid conjugates were synthesized and tested for their inhibitory effects on N-glycan specific ST6GAL1 and O-glycan specific ST3GAL1. The LCA-amino acid conjugates with phenyl and indole moieties showed enhanced inhibitory activity and selectivity towards the N-glycan-specific ST6GAL1, with the indole-containing compound 4e exhibiting the highest activity (IC50 = 20.0 ± 0.5 μM). In addition, compound 4e exhibited the highest antimetastatic potential, effectively inhibiting MDA-MB-231 cell migration at non-cytotoxic concentrations. Compound 4e also suppressed tumor growth and metastasis in vivo, attributing to its potential to disrupt integrins sialylation. The conjugate has also demonstrated excellent antiangiogenetic properties in vitro and ex vivo, owing to its ability to downregulate the VEGF/VEGFR2/Akt pathway. Taken together, these findings prove the practicality of employing LCA as a scaffold and aromatic amino acid conjugation in the discovery of novel, potent, and selective ST inhibitors necessary to address abnormal cell surface α-2,6-N-sialylation.
Collapse
Affiliation(s)
- Wei-Sheng Chen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan; Department of Chemistry, National Central University, Taoyuan 320, Taiwan; Biomedical Translational Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Christian Angelo P Concio
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan; Biomedical Translational Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Tzu-Ting Chang
- Biomedical Translational Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chia-Ling Chen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Ser John Lynon P Perez
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan; Biomedical Translational Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Wen-Shan Li
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan; Biomedical Translational Research Center, Academia Sinica, Taipei 115, Taiwan; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; PhD Program in Biotechnology Research and Development, Taipei Medical University, Taipei 115, Taiwan; Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
6
|
Xu N, Li Z, Zeng X, Jiang Y, Sun T, Liu S, Li N, Li Z, Huang Y, Wang L. Reln-Dab1 pathway mitigates retinal ganglion cell apoptosis in retinal ischemia-reperfusion injury. Cell Death Dis 2025; 16:423. [PMID: 40442071 PMCID: PMC12122947 DOI: 10.1038/s41419-025-07742-6] [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: 08/28/2024] [Revised: 05/06/2025] [Accepted: 05/19/2025] [Indexed: 06/02/2025]
Abstract
Ischemia-reperfusion (I/R) injury is associated with a variety of retinal diseases, resulting in loss of the number of ganglion cells (RGCs), retinal structural disorders, and retinal dysfunction. The Reelin protein is an important regulator of neuronal migration and synaptogenesis, and the Reln signaling pathway plays an essential role in regulating the targeted projection of RGC dendrites and neuronal survival, which has not been reported in retinal I/R injury. The aim of this study was to investigate the expression, role and mechanism of Reln in retinal I/R injury. By establishing Reln-CreERT2 mTmG transgenic mice, it was observed that the expression of Reln initially decreased and then increased after retinal I/R injury. After supplementing exogenous Reelin protein and adeno-associated virus (AAV)-targeted regulation of Reln in vivo, morphological and functional experiments demonstrated its effectiveness in protecting RGCs survival, maintaining morphological integrity of the retina, and inhibiting post-injury retinal dysfunction. Furthermore, integrin β1 (Itgb1) was identified as the main receptor through which Reelin exerts neuroprotective effects while regulating retinal I/R injury repair through the Dab1-PI3K-Akt pathway. These findings provide evidence supporting Reln pathway's role in maintaining retinal homeostasis and facilitating injury repair. Moreover, these findings have significant implications for identifying new targets for preventing and treating various retinal diseases.
Collapse
Affiliation(s)
- Ning Xu
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Zongyuan Li
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Xiangwen Zeng
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Yilin Jiang
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Tunan Sun
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Shuyu Liu
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Na Li
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Zhao Li
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Yifei Huang
- Medical School of Chinese People's Liberation Army, Beijing, China.
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China.
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China.
| | - Liqiang Wang
- Medical School of Chinese People's Liberation Army, Beijing, China.
- Department of Ophthalmology, The Third Medical Center, General Hospital of Chinese People's Liberation Army, Beijing, China.
- State Key Laboratory of Kidney Diseases, General Hospital of Chinese People's Liberation Army, Beijing, China.
| |
Collapse
|
7
|
Ishiyama T, Sano D, Takahashi H, Oridate N, Suzuki Y, Fujii S. Ligand-Receptor Interactions Between Squamous and Endothelial Cells Induce Head and Neck Squamous Cell Carcinoma. Cancer Sci 2025. [PMID: 40418639 DOI: 10.1111/cas.70085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/28/2025] Open
Abstract
Advances in narrowband imaging (NBI) have revealed that squamous epithelial lesions form alongside changes in squamous epithelial cells and intrapapillary capillary loops (IPCLs) in the head and neck. However, the molecular interactions between squamous epithelial cells and endothelial cells (ECs) that promote IPCL proliferation are unclear. This study aimed to identify the mechanisms of cooperation between parenchymal squamous cells and stromal IPCLs during the formation of head and neck squamous cell carcinoma (SCC). We investigated ligand-receptor interactions between squamous epithelial and endothelial cells of IPCLs using Visium analysis on frozen, formalin-fixed and paraffin-embedded (FFPE) tissues from hypopharyngeal squamous epithelial lesions. We examined the protein expression in hypopharyngeal superficial squamous epithelial lesions using immunohistochemistry and immunofluorescence. mRNA expression levels of these genes in SCC and non-tumor tissues were analyzed using RT-qPCR. Phenotypic changes were analyzed by inducing candidate genes into SCC cell lines via a lentivirus system. Visium analysis revealed that Fibronectin 1 (FN1) acted as a ligand in endothelial cells, Cellular communication network factor 1 (CCN1) as a ligand in SCC cells, and Integrin subunit alpha V (ITGAV) as a receptor for both FN1 and CCN1. The expression of these three candidates increased in low-grade dysplasia, an early stage of neoplastic lesions, and was significantly higher in invasive SCCs, except for CCN1. When ITGAV was introduced into SCC cell lines (FaDu and Detroit 562) and HaCaT cells treated with FN1, the cells showed increased proliferation ability. SCC develops via ligand-receptor molecular interactions between squamous epithelial and vascular endothelial cells in IPCLs.
Collapse
Affiliation(s)
- Takahiro Ishiyama
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Daisuke Sano
- Department of Otolaryngology Head and Neck Surgery, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Hideaki Takahashi
- Department of Otolaryngology Head and Neck Surgery, Yokohama City University, Yokohama, Kanagawa, Japan
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Nobuhiko Oridate
- Department of Otolaryngology Head and Neck Surgery, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Satoshi Fujii
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
8
|
Tai L, Zhu D, Tang P, Li J, Li J, Li P, Tao Z, Lei H, Miao K, Wang HX, Lin S, Zhang L, Dou M, Han Y, Shen HM, Deng C, Wang L, Di LJ. Reciprocal stabilization of CtBP and TRIM28 represses autophagy to promote metastasis. Nat Struct Mol Biol 2025:10.1038/s41594-025-01554-0. [PMID: 40374929 DOI: 10.1038/s41594-025-01554-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/05/2025] [Indexed: 05/18/2025]
Abstract
Deciphering the processes through which cancer cells overcome stress, escape a repressive microenvironment and metastasize remains a challenge. Autophagy has been demonstrated to regulate cancer metastasis and C-terminal binding protein (CtBP) has been previously implicated in promoting metastasis in breast cancer. Here we identify the formation of a complex between CtBP and tripartite-motif-containing protein 28 (TRIM28) in the nucleus. Interestingly, this complex regulates the stability of both proteins, as the removal of either partner leads to degradation of the other. Furthermore, the stability of this complex in the nucleus inhibits autophagy through two independent mechanisms. Firstly, the formation of the complex sequesters TRIM28 in the nucleus, preventing its involvement in and its degradation through autophagy. Secondly, this complex participates in the suppression of PTEN expression and leads to inhibition of Unc-51-like kinase 1-mediated autophagy through activation of the protein kinase B-mammalian target of rapamycin pathway. Using mammary gland-specific CtBP-knockout mice, we demonstrate that repression of autophagy by the CtBP-TRIM28 complex modulates luminal duct formation. In breast cancer models, CtBP-TRIM28-dependent inhibition of cellular autophagy also promotes malignant metastasis. Therefore, our study reveals similarities between the mechanisms driving tumor progression and those involved in normal mammary gland development, potentially helping to pave the way toward targeted intervention in breast cancer metastasis.
Collapse
Affiliation(s)
- Lixin Tai
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Dongliang Zhu
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Ping Tang
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Jiajia Li
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Junyi Li
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Peipei Li
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Zhonghua Tao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haipeng Lei
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Kai Miao
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Hong-Xia Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuhai Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Science, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Lei Zhang
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, China
| | - Man Dou
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Metabolomics core, Faculty of Health Sciences, University of Macau, Macau, China
| | - Yu Han
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Genomics, Bioinformatics and Single Cell Analysis Core, Faculty of Health Sciences, University of Macau, Macau, China
| | - Han-Ming Shen
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Chuxia Deng
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
| | - Li Wang
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China
- Metabolomics core, Faculty of Health Sciences, University of Macau, Macau, China
| | - Li-Jun Di
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macau, China.
- Ministry of Education Frontiers Science Center for Precision Oncology (FSCPO), University of Macau, Macau, China.
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, China.
| |
Collapse
|
9
|
Wang M, Guo Z, Zhao S, Liu L, Shi Y, Li H, Su J, Zhang N, Li J, Wu Y. CD49d promotes T-cell senescence in chronic lymphocytic leukaemia. Br J Haematol 2025. [PMID: 40375447 DOI: 10.1111/bjh.20135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/25/2025] [Indexed: 05/18/2025]
Abstract
While CD49d (α4 integrin) is an established prognostic marker in chronic lymphocytic leukaemia (CLL) and is associated with aggressive disease, its impact on T-cell biology remains poorly understood. Compared to healthy donors, CLL patients exhibited significantly elevated CD49d expression in both CD4+ and CD8+ T cells (p < 0.001) as detected by flow cytometry, which was also confirmed by the single-cell RNA sequencing (scRNA-seq) (p < 0.001). Differentially expressed genes in CD49d+ T (both CD8+ and CD4+ T cells) versus CD49d- T cells identified in CLL patients were enriched in cellular senescence pathways, while this phenomenon is absent in healthy individuals. Functional validation demonstrated that CD49d+ T cells displayed elevated senescence-associated markers (e.g. interferon-gamma, granzyme B) and a shift towards memory phenotypes, correlating with immunosuppressive signatures. This discovery suggests that targeting CD49d-dependent senescence pathways may reverse T-cell dysfunction in CLL immunotherapy.
Collapse
Affiliation(s)
- Min Wang
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Zhen Guo
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Sishu Zhao
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Lu Liu
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Yu Shi
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Hui Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Su
- Department of Hematology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Ninghan Zhang
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Jianyong Li
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Yujie Wu
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
He LM, Borjigin S, Chen XQ, Yan ZL, Wang MJ. Therapeutic potential of integrins in diabetic retinopathy. World J Diabetes 2025; 16:101509. [DOI: 10.4239/wjd.v16.i5.101509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/17/2025] [Accepted: 02/20/2025] [Indexed: 04/25/2025] Open
Abstract
Diabetic retinopathy (DR), a leading cause of visual loss, is the result of microvascular damage induced by prolonged hyperglycemia. Numerous studies have revealed the pivotal role of integrins in the pathogenesis of DR, particularly in key processes such as inflammation, vascular leakage, microthrombus formation, and angiogenesis. Consequently, targeting integrins is considered a promising strategy for the treatment of DR. This review focuses on the function of integrins in DR and their potential as therapeutic targets. It describes the molecular mechanisms through which integrins influence DR progression and summarizes the latest outcomes of integrin antagonist-based therapeutic strategies in clinical studies, evaluating their efficacy and potential challenges, which offer promise as novel treatment options for DR.
Collapse
Affiliation(s)
- Li-Mei He
- Department of Internal Medicine, Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Sarul Borjigin
- Department of Endocrinology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Xin-Qi Chen
- Department of Internal Medicine, Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Zhao-Li Yan
- Department of Endocrinology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Ming-Jie Wang
- Department of Endocrinology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
| |
Collapse
|
11
|
Ge Y, Zhu X, Zhang Z, Zhu H, Wang T, Cui J, Zeng H, Wu X, Zhang Z. RGD peptide/dextran sulfate-based nanocarriers loaded with triptolide for double-targeted apoptosis of both tumor cells and M2-like TAMs in pancreatic cancer therapy. Int J Biol Macromol 2025; 311:144032. [PMID: 40345287 DOI: 10.1016/j.ijbiomac.2025.144032] [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: 01/09/2025] [Revised: 04/08/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Triptolide (TP) is a promising anti-tumor candidate derived from the herb Tripterygium wilfordii, but its poor water solubility and multi-organ toxicity limit its application. Here, we developed novel nanoparticles (TP-SP@NPs) modified with dextran sulfate and RGD peptide for double-targeted delivery of TP to both tumor cells and pro-tumor macrophages in pancreatic cancer treatment. TP-SP@NPs exhibited suitable particle size (about 98 nm), good stability and controlled release performance. TP-SP@NPs showed high cellular uptake in Pan02 cells and M2 macrophages through αvβ3 integrin-RGD interaction and SR-A-DS interaction, effectively inhibiting tumor growth by triggering apoptosis of these cells. In Pan02 tumor-bearing mice, TP-SP@NPs specifically accumulated at the tumor site and efficiently decreased the number of M2 macrophages, thereby exerting better curative effect on pancreatic cancer and lower systemic toxicity as compared with TP. As a result, TP-SP@NPs had achieved selective anti-tumor effect, good biosafety and great promise in clinical application.
Collapse
Affiliation(s)
- Yaning Ge
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China
| | - Xin Zhu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China
| | - Zhengxian Zhang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China
| | - Huanhuan Zhu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China
| | - Tianqi Wang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province 450046, PR China
| | - Jingru Cui
- Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China
| | - Huahui Zeng
- Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province 450046, PR China.
| | - Xiangxiang Wu
- Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province 450046, PR China.
| | - Zhenqiang Zhang
- Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, PR China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province 450046, PR China.
| |
Collapse
|
12
|
Huang H, Zhang W, Wu Q, Zhang L, Wu Y, Tong H, Su M. Fucoxanthin Targets β1 Integrin to Disrupt Adhesion and Migration in Human Glioma Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:10961-10973. [PMID: 40261208 DOI: 10.1021/acs.jafc.4c10108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Glioblastoma, the most aggressive type of primary brain tumor, is marked by high invasiveness and metastasis, posing significant challenges in treatment. Fucoxanthin, a carotenoid derived from brown macroalgae, has demonstrated therapeutic potential in cancer therapy; however, its precise mechanisms of action remain unclear. In this study, we explored the inhibitory effects of fucoxanthin on integrin-mediated adhesion and migration in human glioma U-87 MG cells, shedding light on its potential antimetastatic properties. Our data indicated that fucoxanthin at 1 μM did not affect cell viability but inhibited integrin-mediated adhesion of human glioma U-87 MG cells to fibronectin, a key extracellular matrix (ECM) ligand for integrins, without affecting adhesion to poly-l-lysine, a nonintegrin ligand, indicating its selective impact on integrin-mediated adhesion. Fucoxanthin treatment significantly reduced the size and number of focal adhesions (FA), which play a central role in cell adhesion and migration. In addition, fucoxanthin significantly impaired U-87 MG cell migratory capacity, including a reduced accumulated migration distance and velocity, determined by time-lapse videomicroscopy. Further, fucoxanthin remarkably inhibited integrin engagement-mediated actin polymerization, Vav3 phosphorylation, and the downstream activation of Rac1, FAK, and paxillin, further supporting its role in disrupting integrin signaling and cytoskeletal remodeling. Additionally, complementary experiments utilizing protein binding assays, competitive ELISA, CETSA, DARTS, and MST collectively confirmed the direct interaction between fucoxanthin and β1 integrin as well as reduced ligand affinity of β1 integrin for fibronectin. The theoretical model of molecular docking and the dynamics simulation align with our experimental findings, providing a plausible mechanism by which fucoxanthin competitively inhibits the binding of β1 integrin to fibronectin. In summary, our study highlights fucoxanthin as a promising therapeutic agent that impairs integrin-mediated adhesion and migration in glioblastoma cells by directly targeting β1 integrin and disrupting integrin signaling pathways. These findings offer valuable insights into the potential of fucoxanthin as an antimetastatic agent in glioblastoma treatment.
Collapse
Affiliation(s)
- Hui Huang
- Department of Pharmacy, Wenzhou Hospital of Intergrated Traditonal Chinese and Western Medicine, Wenzhou 325000, China
| | - Wen Zhang
- Department of Neurosurgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Qifang Wu
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Lin Zhang
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Yu Wu
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Haibin Tong
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing 100700, China
| | - Meng Su
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| |
Collapse
|
13
|
Song Y, Xu T, Li H, Liu J, Cao S, Yang Y, Li N, Lv P, Han M, Sun H, Dang G, Li J, Sun H, Xin T, Xia H, Zhang C. Delivery of Itgb1-siRNA by triptolide-modified and anti-Flt1 peptide-guided ionizable cationic LNPs for targeted therapy of corneal neovascularization. J Control Release 2025; 383:113811. [PMID: 40324532 DOI: 10.1016/j.jconrel.2025.113811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 04/10/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Corneal neovascularization (CoNV) is a leading cause of visual impairment worldwide. However, CoNV remains challenging to cure clinically because of the limitations of current drugs. New and more effective therapeutic formulations for CoNV treatment are therefore urgently needed. Antisense oligonucleotide drugs hold great promise for the treatment of neovascular diseases, and ionizable cationic lipid nanoparticles (icLNPs) have shown excellent performance for nucleic acid delivery, with high encapsulation, good cellular uptake, and effective endosomal escape. In the present study, we identified integrin β1 (Itgb1) as a key gene involved in angiogenesis and revealed the significant upregulation of Flt1 in vascular endothelial cells and pericytes in CoNV using single-cell sequencing. Itgb1 knockdown significantly inhibited the proliferation and migration of vascular endothelial cells and CoNV in mice. Based on these findings, we developed Itgb1-small interfering RNA (siRNA)-loaded icLNPs, and conjugated anti-Flt1 peptide to their surface to improve CoNV targeting. Furthermore, because lipid nanoparticles reportedly trigger immune responses by upregulating pro-inflammatory cytokine expression, which may promote neovascularization, we modified triptolide (a compound with anti-inflammatory properties) into the icLNPs. The triptolide-modified, anti-Flt1 peptide-conjugated, and Itgb1-siRNA-loaded icLNPs (Itgb1-siRNA@TPL) effectively inhibited the proliferation and migration of vascular endothelial cells in vitro and CoNV in mice after eye drop administration. These effects occurred via downregulation of the PI3K/AKT and NF-κB signaling pathways. Finally, the biosafety of Itgb1-siRNA@TPL was tested, and the results revealed that it was not toxic to the cornea or major organs and had no impact on corneal epithelial healing. In conclusion, Itgb1-siRNA@TPL represent a novel, noninvasive, and effective approach for the treatment of CoNV.
Collapse
Affiliation(s)
- Yuwen Song
- Department of Ophthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, Shandong, China; Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, Shandong, China
| | - Tingting Xu
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Hao Li
- Department of Ophthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, Shandong, China
| | - Jing Liu
- Department of Ophthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, Shandong, China; Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, Shandong, China
| | - Shumin Cao
- Department of Ophthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, Shandong, China; Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, Shandong, China
| | - Yichen Yang
- Department of Ophthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, Shandong, China; Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, Shandong, China
| | - Nianlu Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan 250014, Shandong, China
| | - Peiwen Lv
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan 250014, Shandong, China
| | - Min Han
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan 250014, Shandong, China
| | - Haohan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan 250014, Shandong, China
| | - Guangfu Dang
- Department of Ophthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, Shandong, China
| | - Jianxin Li
- Department of Ophthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, Shandong, China
| | - Hao Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan 250014, Shandong, China
| | - Tao Xin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan 250014, Shandong, China.
| | - Huitang Xia
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan 250014, Shandong, China.
| | - Canwei Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, Shandong, China; Department of Neurosurgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, Jiangxi, China.
| |
Collapse
|
14
|
Bruschi A, Sambri A, Fiore M, Bubbico E, Scollo C, Pace A, Zunarelli R, Montanari A, Cappelli A, Di Prinzio L, De Paolis M. Inside a Metastatic Fracture: Molecular Bases and New Potential Therapeutic Targets. Cancer Med 2025; 14:e70901. [PMID: 40304052 PMCID: PMC12041892 DOI: 10.1002/cam4.70901] [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: 01/12/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
INTRODUCTION Bone metastases and pathological fractures significantly impact the prognosis and quality of life in cancer patients. However, clinical and radiological features alone have been shown to fail to predict skeletal related events of a bone metastasis (SREs). AIM This study focuses on key molecular players including Matrix Metalloproteinases (MMPs), Integrins, Bone Morphogenetic Proteins (BMPs), Parathormone-related Protein (PTHrP). RESULTS The RANK/RANKL/Osteoprotegerin (OPG) pathway, and N-terminal peptide (NTx), involved in the metastatic process and bone integrity disruption. Elevated levels of these molecules have been pointed out as potential biomarkers for predicting SREs, but they have been poorly investigated. Moreover, batimastat, marimastat, tanomastat, andecaliximab, and HIV protease targeting MMPs; Volociximab/M200, cilengitide, abituzumab, and FAK inhibitors targeting integrins; LDN193189, DMH1, and ISLR modulators targeting BMPs; and PTH (7-33)-CBD targeting PTHrP have shown promising results antagonizing these molecules, but no effect on preventing and managing metastatic fractures has been assessed yet. CONCLUSIONS This paper underscores the importance of advanced molecular biology and transcriptomics in identifying novel therapeutic targets. The integration of these biomarkers with clinical and radiological assessments using artificial intelligence tools could revolutionize the diagnostics and treatment strategies for patients with bone metastases.
Collapse
Affiliation(s)
- Alessandro Bruschi
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
| | - Andrea Sambri
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Michele Fiore
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
- Department of Medical and Surgical SciencesAlma Mater Studiorum University of BolognaBolognaItaly
| | - Elisa Bubbico
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Cristina Scollo
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Andrea Pace
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Renato Zunarelli
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Andrea Montanari
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Alberta Cappelli
- Department of RadiologyIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Lorenzo Di Prinzio
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Massimiliano De Paolis
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| |
Collapse
|
15
|
Yang C, Ma H, Liang Z, Zhuang Y, Hu L, Zhang K, Huang L, Li M, Zhang S, Zhen Y. Cyclic RGD modified dextran-quercetin polymer micelles for targeted therapy of breast cancer. Int J Biol Macromol 2025; 308:142272. [PMID: 40118409 DOI: 10.1016/j.ijbiomac.2025.142272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Quercetin is a natural flavonoid found in many plants which has various pharmacological activities including antitumor effect. However, the poor water solubility and bioavailability limit the potential benefits of quercetin for patients. Thus, modifying quercetin structure and developing actively targeted drug delivery systems are extremely important for tumor precision therapy. Herein, polymer-drug conjugates dextran-quercetin (D-Q) and cRGD-dextran (R-D) were synthesized by grafting quercetin and polypeptide cRGDfk (Arg-Gly-Asp-(D-Phe)-Lys) to dextran. Then cRGD-modified dextran-quercetin polymer micelles (R-D-Q) were constructed by self-assembling of D-Q and R-D. R-D-Q micelles possessed appropriate particle size (133.4 nm), nearly neutral potential (8.14 mV) and excellent drug-loading efficiency (13.1 %) and achieved higher cytotoxicity, apoptosis induction and penetration to human breast cancer MCF-7 cells than the micelles unmodified with cRGD, which were ascribed to cRGD-integrin mediated transcytosis. R-D-Q micelles effectively suppressed tumor growth in tumor-bearing mice by delivering more quercetin throughout the tumor tissue. And R-D-Q micelles could promote the apoptosis of tumor cells by activating p38 and JNK signal pathways and suppressing ERK signal pathway. In addition, R-D-Q micelles had no damage to normal tissues of mice at therapeutic dose. These results indicate promising prospects for R-D-Q micelles as an effective drug delivery system against tumor.
Collapse
Affiliation(s)
- Chunpeng Yang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Huiling Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ze Liang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ying Zhuang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Litao Hu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Kexin Zhang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Leixiao Huang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Min Li
- Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, China; Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, China.
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
16
|
Barhoum M, Brassart‐Pasco S, Dupont‐Deshorgue A, Thierry A, Kanagaratnam L, Brassart B, Ramaholimihaso F, Botsen D, Carlier C, Brugel M, Perrier M, Ramont L, Bouché O. Circulating Exosomal Proteins as New Diagnostic Biomarkers for Colorectal Cancer (EXOSCOL01): A Pilot Case-Controlled Study Focusing on MMP14 Potential. J Clin Lab Anal 2025; 39:e70016. [PMID: 40244893 PMCID: PMC12078757 DOI: 10.1002/jcla.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/18/2025] [Accepted: 02/28/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the second leading cause of cancer-related death worldwide. The French CRC screening campaign is based on fecal immunochemical tests (FIT), confirmed by colonoscopy, an invasive procedure with a poor participation rate. This study aimed to compare the expression of circulating exosomal proteins (MMP14, β1-Integrin subunit, β3-Integrin subunit, and α1(I) Collagen chain) in patients with CRC or adenomas. METHODS A total of 71 patients were recruited, including 24 controls (normal colonoscopy), 11 patients with adenoma, and 36 with CRC. Plasmatic exosomal protein expression was measured by western blot analysis and reported to either protein or exosome content. RESULTS The three groups were comparable regarding clinical characteristics. A significant difference was observed for MMP14 relative expression (p = 0.0007), MMP14 expression reported to exosomal protein content (p = 0.0003), and MMP14 expression reported to exosome content (p = 0.0005). These three parameters were significantly higher in patients with adenoma vs. control patients (p = 0.0013, p = 0.0004, and p = 0.0003, respectively). Only MMP14 relative intensity was significantly higher in the CRC group vs. the control group (p = 0.0018). CONCLUSIONS Exosomal MMP14 is a promising early diagnostic biomarker for CRC and adenoma. These preliminary results warrant confirmation in larger studies using quantitative measurements such as ELISA or flow cytometry.
Collapse
Affiliation(s)
- Mickaël Barhoum
- Department of Gastroenterology and Digestive OncologyUniversité de Reims Champagne‐ArdenneReimsFrance
| | - Sylvie Brassart‐Pasco
- Université de Reims Champagne‐Ardenne, CNRSMEDyC Research Unit, UMR CNRS/URCA 7369ReimsFrance
| | | | - Aurore Thierry
- CHU Reims, Unité d'Aide MéthodologiqueUniversité de Reims Champagne‐Ardenne, VieFra UR 3797ReimsFrance
| | - Lukshe Kanagaratnam
- CHU Reims, Unité d'Aide MéthodologiqueUniversité de Reims Champagne‐Ardenne, VieFra UR 3797ReimsFrance
| | - Bertrand Brassart
- Université de Reims Champagne‐Ardenne, CNRSMEDyC Research Unit, UMR CNRS/URCA 7369ReimsFrance
| | - Fidy Ramaholimihaso
- Department of Gastroenterology and Digestive OncologyUniversité de Reims Champagne‐ArdenneReimsFrance
| | - Damien Botsen
- Department of Medical OncologyGodinot Cancer InstituteReimsFrance
| | - Claire Carlier
- Department of Medical OncologyGodinot Cancer InstituteReimsFrance
| | - Mathias Brugel
- Gastroenterology and Digestive Oncology DepartmentCentre Hospitalier Côte BasqueBayonneFrance
| | - Marine Perrier
- Department of Gastroenterology and Digestive OncologyUniversité de Reims Champagne‐ArdenneReimsFrance
| | - Laurent Ramont
- Université de Reims Champagne‐Ardenne, CNRSMEDyC Research Unit, UMR CNRS/URCA 7369ReimsFrance
- Biochemistry, Pharmacology and Toxicology DepartmentCHU ReimsReimsFrance
| | - Olivier Bouché
- Department of Gastroenterology and Digestive OncologyUniversité de Reims Champagne‐ArdenneReimsFrance
- Université de Reims Champagne‐Ardenne, CNRSMEDyC Research Unit, UMR CNRS/URCA 7369ReimsFrance
| |
Collapse
|
17
|
Finger AM, Hendley AM, Figueroa D, Gonzalez H, Weaver VM. Tissue mechanics in tumor heterogeneity and aggression. Trends Cancer 2025:S2405-8033(25)00096-2. [PMID: 40307158 DOI: 10.1016/j.trecan.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 03/10/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Tumorigenesis ensues within a heterogeneous tissue microenvironment that promotes malignant transformation, metastasis and treatment resistance. A major feature of the tumor microenvironment is the heterogeneous population of cancer-associated fibroblasts and myeloid cells that stiffen the extracellular matrix. The heterogeneously stiffened extracellular matrix in turn activates cellular mechanotransduction and creates a hypoxic and metabolically hostile microenvironment. The stiffened extracellular matrix and elevated mechanosignaling also drive tumor aggression by fostering tumor cell growth, survival, and invasion, compromising antitumor immunity, expanding cancer stem cell frequency, and increasing mutational burden, which promote intratumor heterogeneity. Delineating the molecular mechanisms whereby tissue mechanics regulate these phenotypes should help to clarify the basis for tumor heterogeneity and cancer aggression and identify novel therapeutic targets that could improve patient outcome. Here, we discuss the role of the extracellular matrix in driving cancer aggression through its impact on tumor heterogeneity.
Collapse
Affiliation(s)
- Anna-Marie Finger
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA 94143; Current address: Liver Disease Research, Global Drug Discovery, Novo Nordisk A/S, Malov, Denmark
| | - Audrey Marie Hendley
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA 94143
| | - Diego Figueroa
- Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Hugo Gonzalez
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA 94143; Current address: Laboratory of Tumor Microenvironment and Metastasis, Centro Ciencia & Vida, Santiago, Chile
| | - Valerie Marie Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA 94143; Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
18
|
Gao T, Maskalenko NA, Kabir S, Campbell KS, Wu J. Molecular basis of β2 integrin activation by talin unveils subunit-specific mechanisms of integrin signaling. Cell Rep 2025; 44:115607. [PMID: 40310722 DOI: 10.1016/j.celrep.2025.115607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/07/2025] [Accepted: 04/02/2025] [Indexed: 05/03/2025] Open
Abstract
Integrins consist of 24 species, each with unique tissue expression profiles and distinct biological functions. The β subunit of integrin interacts with the FERM-folded head domain of talin through an NPxY/F motif, triggering integrin activation. Although this motif is conserved across most integrin-β subunits, the precise molecular mechanism governing talin's selective recognition of different integrin-β subunits remains unclear. We identify two distinct configurations of the talin head when interacting with β2 and β3 integrins, providing critical insights into subunit-specific recognition of integrins. Structural studies reveal that mutations at the subdomain interface of the talin head can shift its β2-bound configuration to a β3-bound configuration. This shift enhances β2-integrin affinity, leading to increased lymphocyte function-associated antigen-1 (LFA-1)-mediated natural killer cell activity. Together, our data elucidate the structural basis of talin's role in mediating integrin activation in a subunit-specific manner and advance our understanding of how talin may regulate diverse functions of various integrin species.
Collapse
Affiliation(s)
- Tong Gao
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Nicholas A Maskalenko
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Salvin Kabir
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Department of Biology, College of Science & Technology, Temple University, Philadelphia, PA 19122, USA
| | - Kerry S Campbell
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Jinhua Wu
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| |
Collapse
|
19
|
Terrell JA, Chen C. Extracellular Matrix Microstructures Modulate Hepatic Methionine Cycle and Methylations. Biomacromolecules 2025. [PMID: 40298277 DOI: 10.1021/acs.biomac.4c01748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The field of mechanobiology has grown in the past decade, but limited studies investigate how the extracellular matrix affects the cell metabolome. The methionine cycle involves the catabolism and regeneration of methionine through the donation and recovery of a single methyl group; this methyl group can methylate DNA, RNA, and proteins to alter gene expression and protein-protein interactions. Through studying cells cultured on fibrous (mimicking healthy extracellular matrice (ECM)) and flat (mimicking severely fibrotic ECM) substrates, we observed an increase in methionine cycle enzyme expression in cells on the flat substrate. We also present how the methionine cycle is modulated by the ECM through transmembrane protein integrin β1. By inhibiting integrin activation through the ligand-mimicking peptide RGD, we observed that the methionine cycle was protected from alteration. The results presented provide insight into possible therapeutic targets for fibrotic diseases and knowledge of mechanisms by which the ECM alters cell processes.
Collapse
Affiliation(s)
- John A Terrell
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County, Baltimore, Maryland 21250, United States
| | - Chengpeng Chen
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County, Baltimore, Maryland 21250, United States
| |
Collapse
|
20
|
Stawarska A, Bamburowicz-Klimkowska M, Bystrzejewski M, Kasprzak A, Grudzinski IP. Carbon-Encapsulated Iron Nanoparticles Seeking Integrins in Murine Glioma. Int J Nanomedicine 2025; 20:5475-5488. [PMID: 40321805 PMCID: PMC12048782 DOI: 10.2147/ijn.s511286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Purpose Targeting integrin receptors for MRI represents a novel method in diagnosing glioblastoma. In the present study carbon-encapsulated iron nanoparticles to explore murine glioma tracking based upon specific direct targeting with monoclonal antibodies against the beta-3 subunit (CD61) of the integrin αVβ3 receptor are described. Methods The carbon arc discharge method was used to synthesize nanoparticles and amidation-type reaction were applied to attach monoclonal antibody (anti-CD61) with acidic group functionalized nanoparticles to lead two types of bioconjugates (Fe@C-CONH-anti-CD61 and Fe@C-(CH2)2-CONH-anti-CD61). The as-synthesized bioconjugates were tested on murine glioma cells (GL261) using MTT, LDH and calcein AM/propidium iodide assays. Relaxometry measurements were performed with a 1.5 T (63 MHz) MRI scanner using both GL261 cells and C57BL/6 mice bearing GL261 tumors. Results The results showed that Fe@C-CONH-anti-CD61 and Fe@C-(CH2)2-CONH-anti-CD61 nanoparticles have higher binding affinity towards GL261 cells compared to pristine nanoparticles without antibodies. Studies evidenced that the antibody-decorated nanoparticles did not produce any severe cytotoxic effects on murine glioma cells. Preclinical MRI studies demonstrated that the Fe@C-(CH2)2-CONH-anti-CD61 nanoparticle-based construct specifically targeted murine glioma in animals. Conclusion The carbon-encapsulated iron nanoparticles functionalized with monoclonal antibodies recognizing the beta-3 subunit of the integrin αVβ3 receptor can be considered as a potential contrast agent for MRI-based tracking glioblastoma.
Collapse
Affiliation(s)
- Agnieszka Stawarska
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | | | - Michał Bystrzejewski
- Department of Physical Chemistry, Faculty of Chemistry, Warsaw University, Warsaw, Poland
| | - Artur Kasprzak
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Ireneusz P Grudzinski
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
21
|
Ding X, Liang Y, Zhou S, Wu Y, Sałata P, Mikolajczk-Martinez A, Khosrawipour V, Zhang Z. Targeting tumor extracellular matrix with nanoparticles to circumvent therapeutic resistance. J Control Release 2025; 383:113786. [PMID: 40306575 DOI: 10.1016/j.jconrel.2025.113786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
Each stage of tumor development is intrinsically linked to the tumor microenvironment (TME), wherein the extracellular matrix (ECM) serves as a vital and abundant component in tumor tissues. The ECM is a non-cellular, three-dimensional macromolecular network scaffold that provides structural support to cells, stores bioactive molecules, and mediates signaling pathways through specific binding to cell surface receptors. Moreover, the ECM in tumor tissues plays a crucial role in impeding drug diffusion and resisting apoptosis induced by conventional anti-cancer therapies that primarily target cancer cells. Therefore, directing attentions towards the tumor ECM can facilitate the identification of novel targets and the development of new therapies. This review aims to summarize the composition, structure, remodeling, and function of tumor ECM, its association with drug resistance, and current targeting strategies, with a specific emphasis on nanoparticles (NPs).
Collapse
Affiliation(s)
- Xinyue Ding
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Yiyu Liang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Siyuan Zhou
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Yao Wu
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Patricia Sałata
- Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | | | | | - Zhiwen Zhang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China.
| |
Collapse
|
22
|
Sankar S, Kalidass B, Indrakumar J, Kodiveri Muthukaliannan G. NSAID-encapsulated nanoparticles as a targeted therapeutic platform for modulating chronic inflammation and inhibiting cancer progression: a review. Inflammopharmacology 2025:10.1007/s10787-025-01760-8. [PMID: 40285986 DOI: 10.1007/s10787-025-01760-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025]
Abstract
Recent advancements in nanotechnology have significantly advanced nanocarrier-mediated drug delivery systems, promoting therapeutic outcomes in mitigating chronic inflammation and cancer. Nanomaterials offer significant advantages over traditional small-molecule drugs, including a high surface-area-to-volume ratio, tunable structural features, and extended bloodstream circulation time. Chronic inflammation is a well-established mechanism for malignant initiation, progression, and metastasis, promoting the potent strategy for cancer prevention and therapy. Numerous studies revealed that nonsteroidal anti-inflammatory drugs (NSAIDs) have the therapeutic ability to manage disease progression via amolerating angiogenesis and inducing apoptosis. However, prolonged intake of NSAIDs is often limited by adverse side-effects and systemic toxicities. The encapsulation of NSAIDs in a nanocarrier have materialized as a dynamic approach to mitigate the limitations by improving pharmacokinetics and pharmacodynamics, reducing off-target effects, and enhancing the drug stability. This review encompasses recent progress in the development of NSAID-based nanotherapeutics, focusing on pivotal mechanisms underlying nanoparticle-mediated drug delivery, such as improved tumor-specific targeting and strategies to overcome drug resistance. The ability of these nano-cargoes to accommodate anti-inflammatory strategies with advanced drug delivery platforms is critically evaluated. This review also highlights the transformative potential of NSAID-encapsulated nanoparticles as a multifaceted therapeutic venue for addressing chronic inflammation and mitigating cancer progression.
Collapse
Affiliation(s)
- Srivarshini Sankar
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632 014, India
| | - Bharathi Kalidass
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632 014, India
| | - Janani Indrakumar
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632 014, India
| | - Gothandam Kodiveri Muthukaliannan
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632 014, India.
| |
Collapse
|
23
|
Boutelle AM, Mabene AR, Yao D, Xu H, Wang M, Tang YJ, Lopez SS, Sinha S, Demeter J, Cheng R, Benard BA, McCrea EM, Valente LJ, Drainas AP, Fischer M, Majeti R, Petrov DA, Jackson PK, Yang F, Winslow MM, Bassik MC, Attardi LD. Integrative multiomic approaches reveal ZMAT3 and p21 as conserved hubs in the p53 tumor suppression network. Cell Death Differ 2025:10.1038/s41418-025-01513-8. [PMID: 40263541 DOI: 10.1038/s41418-025-01513-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 02/18/2025] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
TP53, the most frequently mutated gene in human cancer, encodes a transcriptional activator that induces myriad downstream target genes. Despite the importance of p53 in tumor suppression, the specific p53 target genes important for tumor suppression remain unclear. Recent studies have identified the p53-inducible gene Zmat3 as a critical effector of tumor suppression, but many questions remain regarding its p53-dependence, activity across contexts, and mechanism of tumor suppression alone and in cooperation with other p53-inducible genes. To address these questions, we used Tuba-seqUltra somatic genome editing and tumor barcoding in a mouse lung adenocarcinoma model, combinatorial in vivo CRISPR/Cas9 screens, meta-analyses of gene expression and Cancer Dependency Map data, and integrative RNA-sequencing and shotgun proteomic analyses. We established Zmat3 as a core component of p53-mediated tumor suppression and identified Cdkn1a as the most potent cooperating p53-induced gene in tumor suppression. We discovered that ZMAT3/CDKN1A serve as near-universal effectors of p53-mediated tumor suppression that regulate cell division, migration, and extracellular matrix organization. Accordingly, combined Zmat3-Cdkn1a inactivation dramatically enhanced cell proliferation and migration compared to controls, akin to p53 inactivation. Together, our findings place ZMAT3 and CDKN1A as hubs of a p53-induced gene program that opposes tumorigenesis across various cellular and genetic contexts.
Collapse
Affiliation(s)
- Anthony M Boutelle
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Aicha R Mabene
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - David Yao
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Haiqing Xu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Mengxiong Wang
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuning J Tang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Steven S Lopez
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Sauradeep Sinha
- Department of Bioengineering, Stanford University Schools of Engineering and Medicine, Stanford, CA, USA
| | - Janos Demeter
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ran Cheng
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Brooks A Benard
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Edel M McCrea
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Liz J Valente
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Heligenics Inc, Las Vegas, NV, USA
| | | | - Martin Fischer
- Computational Biology Group, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Ravindra Majeti
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Dmitri A Petrov
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Peter K Jackson
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Fan Yang
- Department of Bioengineering, Stanford University Schools of Engineering and Medicine, Stanford, CA, USA
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael C Bassik
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
24
|
Gao H, Ma Z, Zhu Z, Yang Z, Chen B, Wu X, Jakobsson V, Deng Y, Wang H, Zhang W, Zhang J. Comparative study of [ 18F]AlF-NOTA-FAPI-RGD and [ 18F]FDG/[ 18F]AlF-NOTA-FAPI-04 PET/CT in renal cell carcinoma. Theranostics 2025; 15:5790-5800. [PMID: 40365277 PMCID: PMC12068302 DOI: 10.7150/thno.113070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 04/05/2025] [Indexed: 05/15/2025] Open
Abstract
Renal cell carcinoma (RCC) is a significant global health concern, and the early diagnosis and accurate staging of clear cell renal cell carcinoma (ccRCC) remain major challenges. [18F]FDG PET/CT is not ideal for diagnosing ccRCC due to the low glucose metabolism potential of cancer cells. Both fibroblast activation protein (FAP) and the angiogenic integrin αvβ3 receptor are closely linked to the pathogenesis and progression of ccRCC. The aim of this study is to evaluate a novel radiopharmaceutical [18F]AlF-NOTA-FAPI-RGD (denoted as [18F]AlF-LNC1007), a dual-targeting heterodimer tracer targeting both FAP and integrin αvβ3, and to compare the diagnostic value of [18F]AlF-LNC1007 with [18F]FDG and [18F]AlF-NOTA-FAPI-04 PET/CT in RCC. Materials and Methods: A total of 35 participants, highly suspected to have RCC, were recruited. [18F]AlF-LNC1007 and [18F]AlF-NOTA-FAPI-04/[18F]FDG scans were performed at least one day apart, and both were completed within one week. The Wilcoxon signed-rank test or paired t-test was used to assess differences in tumor uptake and TBR (tumor-to-background ratio) between [18F]AlF-LNC1007 and the other two imaging agents. The Spearman correlation coefficient was used to evaluate the correlation between tumor uptake and the expression of FAP and αvβ3. Results: The detection rate, sensitivity, and positive predictive value (PPV) of [18F]AlF-LNC1007 for RCC primary lesions were significantly higher than those of [18F]FDG, at 91% vs. 76%, 100% vs. 85%, and 91% vs. 87%, respectively. Obvious advantages were also seen in metastatic lesions at 94% vs. 34%, 94% vs. 29%, and 100% vs. 100%. Compared to [18F]AlF-NOTA-FAPI-04, the corresponding detection rate, sensitivity, and PPV were 98% vs. 90%, 100% vs. 92%, and 98% vs. 98% for primary lesions, and 89% vs. 78%, 89% vs. 93%, and 100% vs. 82% for metastatic lesions. The uptake and TBR of [18F]AlF-LNC1007 in both primary and metastatic lesions were significantly higher than those of [18F]FDG (all P < 0.001). The uptake of [18F]AlF-LNC1007 showed a moderate to high positive correlation with the expression levels of αvβ3 and the combined expression of FAP and αvβ3 (r = 0.756, P = 0.0003; r = 0.678, P = 0.0002) and a low positive correlation with FAP expression alone (r = 0.389, P = 0.014). The uptake of [18F]AlF-NOTA-FAPI-04 showed a low to moderate positive correlation with FAP expression and the combined expression of FAP and αvβ3 (r = 0.570, P = 0.0002; r = 0.408, P = 0.010), and no correlation with αvβ3 expression alone (r = 0.262, P = 0.107). Conclusion: [18F]AlF-LNC1007 demonstrated significantly higher diagnostic efficacies and uptake in primary and metastatic renal cell carcinoma (RCC) compared to FDG PET/CT. Additionally, [18F]AlF-LNC1007 exhibited higher diagnostic efficacies and uptake in primary RCC than [18F]AlF-NOTA-FAPI-04 PET/CT. While these findings suggest potential diagnostic advantages, further studies are needed to fully evaluate its diagnostic efficacy compared to the standard of treatment.
Collapse
Affiliation(s)
- Haiyan Gao
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Zhiwei Ma
- Department of Urology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Ziyang Zhu
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Zhichuan Yang
- Department of Emergency Surgery, The Affiliated Chengdu 363 Hospital of Southwest Medical University, Chengdu 610041, China
| | - Bo Chen
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xiaoming Wu
- College of Nuclear Science and Technology, Harbin Engineering University, Harbin 150001, China
| | - Vivianne Jakobsson
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yujiao Deng
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Hao Wang
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Wei Zhang
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
25
|
Zhang Q, Yu S, He S, He Y, Liu X, Wang F. On-Demand Regulation of Catalytic DNA Circuits Using Phosphorylated Charge Reversal Peptides. Angew Chem Int Ed Engl 2025:e202425113. [PMID: 40249733 DOI: 10.1002/anie.202425113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/26/2025] [Accepted: 04/11/2025] [Indexed: 04/20/2025]
Abstract
Catalytic DNA circuits have emerged as a powerful tool for high-performance biosensing application; however, the establishment of a safe and efficient in vivo delivery system remains a critical bottleneck. Peptides serve as attractive carriers due to their rich chemical diversity, excellent biocompatibility, high loading capacity, and specific binding ability, making them ideal candidates for the on-demand regulation of DNA circuits-yet remains largely unexplored. In this study, we developed a multifunctional enzyme-responsive peptide (ERP) for the efficient loading and specific intracellular delivery and release of catalytic circuitry probes through a phosphorylation-based charge reversal procedure. This ERP-programmed catalytic DNA circuit enables the precise, spatially controllable in vivo imaging of microRNA (miRNA). The multifunctional cationic peptide formed a stable nanocomplex with anionic DNA cargo via strong electrostatic interactions, thus protecting the DNA probes from degradation in biological environments. Moreover, with the ability to actively targeting tumor cells and facilitate endogenous phosphorylation-guided release of DNA probes, this multifunctional peptide could significantly reduce the nonspecific delivery of probes to healthy tissues, thereby minimizing unwanted off-site signal leakage. By the integration of cell-selective delivery and site-specific stimulation, this endogenously regulated and multiply guaranteed DNA circuit system paves a simple yet effective way for disease diagnosis.
Collapse
Affiliation(s)
- Qingqing Zhang
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430072, P.R. China
| | - Shanshan Yu
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430072, P.R. China
| | - Shizhen He
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430072, P.R. China
| | - Yuqiu He
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430072, P.R. China
| | - Xiaoqing Liu
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430072, P.R. China
- Research Institute of Shenzhen, Wuhan University, Shenzhen, 518057, P.R. China
| | - Fuan Wang
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430072, P.R. China
- Research Institute of Shenzhen, Wuhan University, Shenzhen, 518057, P.R. China
| |
Collapse
|
26
|
Zheng X, Wu H, Zhu W. SPOCK2 promotes the invasion and migration of ovarian cancer cells through FAK signaling pathway. J Gynecol Oncol 2025; 36:36.e98. [PMID: 40350704 DOI: 10.3802/jgo.2025.36.e98] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 02/09/2025] [Accepted: 03/10/2025] [Indexed: 05/14/2025] Open
Abstract
OBJECTIVE Ovarian cancer is one of the most prevalent malignancies worldwide, with the highest mortality rate among gynecological cancers. This study aims to investigate the molecular mechanisms of SPOCK2 in ovarian cancer progression and metastasis and evaluate its potential as a therapeutic target. METHODS The expression levels of SPOCK2 in ovarian cancer tissues and normal tissues were analyzed using data from The Cancer Genome Atlas (TCGA) and immunohistochemistry experiments. Functional assays, including epithelial-mesenchymal transition (EMT), invasion, and migration assays, were performed in high-grade serous ovarian cancer (HGSOC) cells to explore the role of SPOCK2. The interaction between SPOCK2 and ITGA3 and the subsequent activation of focal adhesion kinase (FAK) signaling were investigated. In vivo experiments were conducted to validate the effects of SPOCK2 knockdown on tumor metastasis and invasiveness. RESULTS SPOCK2 expression was significantly upregulated in ovarian cancer tissues compared to normal tissues and was associated with poor prognosis. Functional assays demonstrated that SPOCK2 promotes EMT, invasion, and migration in HGSOC cells by interacting with ITGA3 and activating FAK signaling. In vivo experiments confirmed that SPOCK2 knockdown significantly suppressed tumor metastasis and invasiveness. CONCLUSION This study highlights the critical role of the SPOCK2/ITGA3 axis in driving ovarian cancer progression and provides evidence for SPOCK2 as a potential molecular marker and therapeutic target. These findings offer new insights into the early diagnosis and treatment of ovarian cancer, with significant clinical implications for improving patient outcomes.
Collapse
Affiliation(s)
- Xiaoli Zheng
- The Second Affiliated Hospital of Soochow University, Suzhou 215000, China
- The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Hua Wu
- The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Weipei Zhu
- The Second Affiliated Hospital of Soochow University, Suzhou 215000, China.
| |
Collapse
|
27
|
Bretl M, Cheng L, Kendziorski C, Thibeault SL. RNA-sequencing demonstrates transcriptional differences between human vocal fold fibroblasts and myofibroblasts. BMC Genomics 2025; 26:347. [PMID: 40197133 PMCID: PMC11974177 DOI: 10.1186/s12864-025-11533-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Differentiation of fibroblasts into myofibroblasts is necessary for wound healing, but excessive myofibroblast presence and persistence can result in scarring. Treatment for scarring is limited largely due to a lack of comprehensive understanding of how fibroblasts and myofibroblasts differ at the transcript level. The purpose of this study was to characterize transcriptional profiles of injured fibroblasts relative to normal fibroblasts, utilizing fibroblasts from the vocal fold as a model. RESULTS Utilizing bulk RNA sequencing technology, we identified differentially expressed genes between four cell lines of normal fibroblasts (cVFF), one line of scarred fibroblasts (sVFF), and four lines of fibroblasts treated with transforming growth factor-beta 1 (TGF-β1), representing an induced-scar phenotype (tVFF). Principal component analysis revealed clustering of normal fibroblasts separate from the clustering of fibroblasts treated with TGF-β1; scarred fibroblasts were more similar to normal fibroblasts than fibroblasts treated with TGF-β1. Enrichment analyses revealed pathways related to cell signaling, receptor-ligand activity, and regulation of cell functions in scarred fibroblasts, pathways related to cell adhesion in normal fibroblasts, and pathways related to ECM binding in fibroblasts treated with TGF-β1. Although transcriptomic profiles between scarred fibroblasts and fibroblasts treated with TGF-β1 were relatively dissimilar, the most highly co-expressed genes were enriched in pathways related to actin cytoskeleton binding, which supports the use of fibroblasts treated with TGF-β1 to represent a scarred cell phenotype. CONCLUSIONS Transcriptomics of normal fibroblasts differ from myofibroblasts, including from those retrieved from scar and those treated with TGF-β1. Despite large differences in transcriptomics between tVFF and sVFF, tVFF serve as a useful in vitro model of myofibroblasts and highlight key similarities to myofibroblasts extracted from scar pathology, as well as expected differences related to normal fibroblasts from healthy vocal folds.
Collapse
Affiliation(s)
- Michelle Bretl
- Department of Communication Sciences and Disorders, University of Wisconsin - Madison, Madison, WI, USA
- Department of Surgery, Division of Otolaryngology, University of Wisconsin - Madison, Madison, WI, USA
| | - Lingxin Cheng
- Department of Biostatistics & Medical Informatics, University of Wisconsin - Madison, Madison, WI, USA
| | - Christina Kendziorski
- Department of Biostatistics & Medical Informatics, University of Wisconsin - Madison, Madison, WI, USA
| | - Susan L Thibeault
- Department of Communication Sciences and Disorders, University of Wisconsin - Madison, Madison, WI, USA.
- Department of Surgery, Division of Otolaryngology, University of Wisconsin - Madison, Madison, WI, USA.
| |
Collapse
|
28
|
Guo S, Sidhu R, Ramar V, Guo AA, Wang G, Liu M. RNA Sequencing Identifies Novel Signaling Pathways and Potential Drug Target Genes Induced by FOSL1 in Glioma Progression and Stemness. Biologics 2025; 19:157-176. [PMID: 40206361 PMCID: PMC11980931 DOI: 10.2147/btt.s509774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/01/2025] [Indexed: 04/11/2025]
Abstract
Background Glioblastoma is a highly aggressive brain tumor, and the transition from the proneural to mesenchymal subtype is associated with more aggressive and therapy-resistant features. However, the signaling pathways and genes involved in this transition remain largely undefined. Methods We utilized patient-derived xenograft (PDX) samples of glioblastoma, specifically PDX-L14, which exhibit both negative and overexpressed FOSL1 expression. mRNA expression profiles were assessed by RNA sequencing in these samples, followed by gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and Gene Set Enrichment Analysis (GSEA). Validation of the hub genes was performed using qPCR and immunohistochemistry assays. Results Differentially expressed genes (DEGs) between FOSL1 overexpression groups were predominantly involved in ferroptosis, immune response, angiogenesis, vascular mimicry, autophagy, epithelial-mesenchymal transition (EMT), cancer cell stemness, temozolomide (TMZ) resistance, and NF-κB signaling. Downregulated DEGs were associated with TMZ resistance, glioma proliferation, RNA processing, and Wnt/β-catenin signaling. Key enrichment pathways, including NF-κB, Want, and BMP, are all critical for maintaining glioma stemness. FOSL1 was found to regulate RNA processing and ubiquitination. Notably, 8 upregulated (ITGA5, SDC1, PHLDB2, TNFRSF8, ADAM8, TLR7, STEAP3, and POU3F2) and 4 downregulated (IFIT1, FBXO16, ARL3, and BEX1) genes were identified, with implications for glioblastoma prognosis. Conclusion This transcriptome investigation emphasizes the diverse functions of FOSL1 in different biological processes and signaling networks during the shift from proneural to mesenchymal state in glioblastoma.
Collapse
Affiliation(s)
- Shanchun Guo
- RCMI Cancer Research Center and Department of Chemistry, Xavier University, New Orleans, LA, USA
| | - Rajveer Sidhu
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Vanajothi Ramar
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Alyssa A Guo
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Guangdi Wang
- RCMI Cancer Research Center and Department of Chemistry, Xavier University, New Orleans, LA, USA
| | - Mingli Liu
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
29
|
Konstantaraki M, Berdiaki A, Neagu M, Zurac S, Krasagakis K, Nikitovic D. Understanding Merkel Cell Carcinoma: Pathogenic Signaling, Extracellular Matrix Dynamics, and Novel Treatment Approaches. Cancers (Basel) 2025; 17:1212. [PMID: 40227764 PMCID: PMC11987840 DOI: 10.3390/cancers17071212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Merkel cell carcinoma (MCC) is a rare but aggressive neuroendocrine skin cancer, driven by either Merkel cell polyomavirus (MCPyV) integration or ultraviolet (UV)-induced mutations. In MCPyV-positive tumors, viral T antigens inactivate tumor suppressors pRb and p53, while virus-negative MCCs harbor UV-induced mutations that activate similar oncogenic pathways. Key signaling cascades, including PI3K/AKT/mTOR and MAPK, support tumor proliferation, survival, and resistance to apoptosis. Histologically, MCC consists of small round blue cells with neuroendocrine features, high mitotic rate, and necrosis. The tumor microenvironment (TME) plays a central role in disease progression and immune escape. It comprises a mix of tumor-associated macrophages, regulatory and cytotoxic T cells, and elevated expression of immune checkpoint molecules such as PD-L1, contributing to an immunosuppressive niche. The extracellular matrix (ECM) within the TME is rich in proteoglycans, collagens, and matrix metalloproteinases (MMPs), facilitating tumor cell adhesion, invasion, and interaction with stromal and immune cells. ECM remodeling and integrin-mediated signaling further promote immune evasion and therapy resistance. Although immune checkpoint inhibitors targeting PD-1/PD-L1 have shown promise in treating MCC, resistance remains a major hurdle. Therapeutic strategies that concurrently target the TME-through inhibition of ECM components, MMPs, or integrin signaling-may enhance immune responses and improve clinical outcomes.
Collapse
Affiliation(s)
- Maria Konstantaraki
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece; (M.K.); (A.B.)
- Dermatology Department, University Hospital of Heraklion, 71110 Heraklion, Greece;
| | - Aikaterini Berdiaki
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece; (M.K.); (A.B.)
| | - Monica Neagu
- Immunology Laboratory, “Victor Babes” National Institute of Pathology, 99-101 Splaiul Independenței, 050096 Bucharest, Romania;
- Pathology Department, Colentina Clinical Hospital, 19-21 Sos Stefan Cel Mare, 020125 Bucharest, Romania;
| | - Sabina Zurac
- Pathology Department, Colentina Clinical Hospital, 19-21 Sos Stefan Cel Mare, 020125 Bucharest, Romania;
- Faculty of Dentistry, University of Medicine and Pharmacy, 8 Eroilor Sanitari Boulevard, 050474 Bucharest, Romania
| | | | - Dragana Nikitovic
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece; (M.K.); (A.B.)
| |
Collapse
|
30
|
Rivas-Mercado E, Neri-Castro E, Zarzosa V, Hernández-Orihuela L, Olvera-Rodríguez F, Torres-Garza JD, Garza-Ocañas L. Mictlan-D3: A novel medium sized RGD-Disintegrin obtained from Crotalus mictlantecuhtli venom, in vitro tested against human breast Cancer and endothelial cells. Toxicol In Vitro 2025; 104:105987. [PMID: 39631634 DOI: 10.1016/j.tiv.2024.105987] [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: 07/19/2024] [Revised: 11/10/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Disintegrins are small non-enzymatic proteins present often at low concentration in the venom of viperid snakes. Isolated disintegrins are known for their lack of toxicity as well as their capacity to antagonize integrin receptors. Integrins are a major family of heterodimeric cell surface receptors that mediate cell-cell and cell-extracellular matrix (ECM) interactions. Integrins regulate key functions in cancer pathology and also tumor development. The aim of this study consisted in the isolation and characterization of disintegrins from rattlesnake new species Crotalus mictlantecuhtli venom. A disintegrin fraction obtained by RP-HPLC and named mictlan-D3, consist in two isoforms of 7439 and 7509 Da with 72 amino acid sequence containing the RGD binding motif. Mictlan-D3 inhibited MDA-MB-231 and HMEC-1 cell adhesion to laminin (LN), fibronectin (FN) and vitronectin (VN), highest inhibition was on MDA-MB-231 cell adhesion to LN by 81 % at 1 μM. The blockade of ⍺Vβ3 integrin was evaluated by wound healing migration assay. Mictlan-D3 inhibited MDA-MB-231 cell migration by 80 % and 38 % after 24 and 72 h of incubation respectively. HMEC-1 cell migration was inhibited by 67.6 % and 27.9 % after 24 and 72 h of incubation. Additionally, mictlan-D3. This work represent the first characterization of disintegrins from the Crotalus mictlantecuhtli venom.
Collapse
Affiliation(s)
- E Rivas-Mercado
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - E Neri-Castro
- Instituto de Biotecnología, Universidad Nacional Autonoma de México, Cuernavaca, Morelos, Mexico
| | - V Zarzosa
- Instituto de Biotecnología, Universidad Nacional Autonoma de México, Cuernavaca, Morelos, Mexico
| | - L Hernández-Orihuela
- Instituto de Biotecnología, Universidad Nacional Autonoma de México, Cuernavaca, Morelos, Mexico
| | - F Olvera-Rodríguez
- Instituto de Biotecnología, Universidad Nacional Autonoma de México, Cuernavaca, Morelos, Mexico
| | - J D Torres-Garza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - L Garza-Ocañas
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico.
| |
Collapse
|
31
|
León-Flores DB, Siañez-Estada LI, Iglesias-Figueroa BF, Siqueiros-Cendón TS, Espinoza-Sánchez EA, Varela-Ramírez A, Aguilera RJ, Rascón-Cruz Q. Anticancer potential of lactoferrin: effects, drug synergy and molecular interactions. Biometals 2025; 38:465-484. [PMID: 40117096 DOI: 10.1007/s10534-025-00672-y] [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: 10/25/2024] [Accepted: 02/19/2025] [Indexed: 03/23/2025]
Abstract
Cancer treatment is among today's most active and challenging research fields. In recent years, significant progress has been made in developing new cancer therapies, including nutraceuticals and natural compounds with anticancer properties. Lactoferrin, a glycoprotein present in mammals, is of significant interest due to its pleiotropic behavior, demonstrating a broad spectrum of biological activities such as antimicrobial, antioxidant, anti-inflammatory, immunomodulatory, and anticancer effects. In this review, we examine the current knowledge of Lf's role in cancer. In addition, it exhibits a synergistic effect along with conventional drugs, potentially enhancing their efficacy and, at the same time, reducing the side effects associated with most traditional therapies. However, it is essential to consider the precise molecular mechanism by which Lf exerts its antitumor activity. Searching interactions with several molecules can provide insight into this mechanism. Additionally, finding lactoferrin receptors can improve the strategies for the specific release of the conjugates. For all these reasons, Lactoferrin becomes a potential therapeutic agent that should be examined in depth.
Collapse
Affiliation(s)
- D B León-Flores
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua. Chihuahua, Chihuahua, México
| | - L I Siañez-Estada
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua. Chihuahua, Chihuahua, México
| | - B F Iglesias-Figueroa
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua. Chihuahua, Chihuahua, México
| | - T S Siqueiros-Cendón
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua. Chihuahua, Chihuahua, México
| | - E A Espinoza-Sánchez
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua. Chihuahua, Chihuahua, México
| | - A Varela-Ramírez
- Border Biomedical Research Center, Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - R J Aguilera
- Border Biomedical Research Center, Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Q Rascón-Cruz
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua. Chihuahua, Chihuahua, México.
| |
Collapse
|
32
|
Zeng C, Chen X, Lin M, Jin Y, Guo Q, Zhou T, Wang X, Li Y, Wang X, Han Y, Du L, Tang Q, Liu P, Zhang J. Overcoming matrix barriers for enhanced immune infiltration using siRNA-coated metal-organic frameworks. Acta Biomater 2025; 196:410-422. [PMID: 40054648 DOI: 10.1016/j.actbio.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 03/16/2025]
Abstract
The extracellular matrix (ECM) of solid tumor constitutes a formidable physical barrier that impedes immune cell infiltration, contributing to immunotherapy resistance. Breast cancer, particularly triple-negative breast cancer (TNBC), is characterized by a collagen-rich tumor microenvironment, which is associated with T cell exclusion and poor therapeutic outcomes. Discoidin domain receptor 2 (DDR2) and integrins, key ECM regulatory receptors on cancer cells, play pivotal role in maintaining this barrier. In this study, we developed a dual-receptor-targeted strategy using metal-organic frameworks (MOFs) to deliver DDR2-specific siRNA (siDDR2) and ITGAV-specific siRNA (siITGAV) to disrupt the ECM barrier. siDDR2 modulates immune infiltration by regulating collagen-cell interactions, while siITGAV suppresses TGF-β1 activation. The MOF@siDDR2+siITGAV complex significantly reduced collagen deposition, enhanced CD8+ T cell infiltration, and downregulated programmed cell death ligand 1 (PD-L1) expression in TNBC. Consequently, this approach markedly inhibited tumor growth. Our findings demonstrate that dual-receptor-targeted MOF-based nanocarriers (MOF@siDDR2+siITGAV) can effectively reprogram the tumor ECM to enhance immune cell access, offering a promising prospect for synergistic cancer immunotherapy. STATEMENT OF SIGNIFICANCE: A dual-receptor-targeted MOF nanocarrier is developed to improve immune accessibility in tumors. Concurrent blockade of DDR2 and ITGAV effectively decreases collagen deposition, increases CD8+ T cell infiltration, and suppresses PD-L1 expression. Modulating the mechanical properties of the extracellular matrix (ECM) to enhance immune accessibility offers an innovative strategy for cancer treatment.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaojing Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China; Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mingxi Lin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yizi Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qing Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Teng Zhou
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xingang Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yiping Li
- Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xinghui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yongming Han
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Ling Du
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Qianyun Tang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| | - Peifeng Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China; Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
33
|
Silver FH. The Role of Connections Between Cellular and Tissue Mechanical Elements and the Importance of Applied Energy in Mechanotransduction in Cancerous Tissue. Biomolecules 2025; 15:457. [PMID: 40305177 PMCID: PMC12025281 DOI: 10.3390/biom15040457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/08/2025] [Accepted: 03/13/2025] [Indexed: 05/02/2025] Open
Abstract
In the presence of cellular mutations and impaired mechanisms of energy transmission to the attached cells and tissues, excess energy is available to upregulate some of the mechanotransduction pathways that maintain cell and tissue structure and function. The ability to transfer applied energy through integrin-mediated pathways, cell ion channels, cell membrane, cytoskeleton-nucleoskeleton connections, cell junctions, and cell-extracellular matrix attachments provides an equilibrium for energy storage, transmission, and dissipation in tissues. Disruption in energy storage, transmission, or dissipation via genetic mutations blocks mechanical communication between cells and tissues and impairs the mechanical energy equilibrium that exists between cells and tissues. This results in local structural changes through altered regulatory pathways, which produce cell clustering, collagen encapsulation, and an epithelial-mesenchymal transition (EMT), leading to increased cellular motility along newly reorganized collagen fibers (fibrosis). The goal of this review is to postulate how changes in energy transfer between cells and the extracellular matrix may alter local energy equilibrium and mechanotransduction pathways. The changes along with cellular mutations lead to cell and ECM changes reported in cancer, which is postulated to modify mechanical equilibria between cells and their ECM. This leads to uncontrolled cancer cellular proliferation and collagen remodeling.
Collapse
Affiliation(s)
- Frederick H Silver
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08854, USA
| |
Collapse
|
34
|
Tiskratok W, Chuinsiri N, Limraksasin P, Kyawsoewin M, Jitprasertwong P. Extracellular Matrix Stiffness: Mechanotransduction and Mechanobiological Response-Driven Strategies for Biomedical Applications Targeting Fibroblast Inflammation. Polymers (Basel) 2025; 17:822. [PMID: 40292716 PMCID: PMC11946729 DOI: 10.3390/polym17060822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
The extracellular matrix (ECM) is a dynamic network providing mechanical and biochemical cues that regulate cellular behavior. ECM stiffness critically influences fibroblasts, the primary ECM producers, particularly in inflammation and fibrosis. This review explores the role of ECM stiffness in fibroblast-driven inflammation and tissue remodeling, focusing on the physicochemical and biological mechanisms involved. Engineered materials, hydrogels, and polydimethylsiloxane (PDMS) are highlighted for replicating tissue-specific stiffness, enabling precise control over cell-matrix interactions. The surface functionalization of substrate materials, including collagen, polydopamine, and fibronectin, enhances bioactivity and fibroblast adhesion. Key mechanotransduction pathways, such as integrin signaling and YAP/TAZ activation, are related to regulating fibroblast behaviors and inflammatory responses. The role of fibroblasts in driving chronic inflammatory diseases emphasizes their therapeutic potentials. Advances in ECM-modifying strategies, including tunable biomaterials and hydrogel-based therapies, are explored for applications in tissue engineering, drug delivery, anti-inflammatory treatments, and diagnostic tools for the accurate diagnosis and prognosis of ECM stiffness-related inflammatory diseases. This review integrates mechanobiology with biomedical innovations, providing a comprehensive prognosis of fibroblast responses to ECM stiffness and outlining future directions for targeted therapies.
Collapse
Affiliation(s)
- Watcharaphol Tiskratok
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (N.C.); (P.J.)
- Oral Health Centre, Suranaree University of Technology Hospital, Nakhon Ratchasima 30000, Thailand
| | - Nontawat Chuinsiri
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (N.C.); (P.J.)
- Oral Health Centre, Suranaree University of Technology Hospital, Nakhon Ratchasima 30000, Thailand
| | - Phoonsuk Limraksasin
- Center of Excellence for Dental Stem Cell Biology, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (P.L.); (M.K.)
| | - Maythwe Kyawsoewin
- Center of Excellence for Dental Stem Cell Biology, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (P.L.); (M.K.)
| | - Paiboon Jitprasertwong
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (N.C.); (P.J.)
- Oral Health Centre, Suranaree University of Technology Hospital, Nakhon Ratchasima 30000, Thailand
| |
Collapse
|
35
|
Huilcaman R, Campos A, Contreras P, Simón L, Varas-Godoy M, Grünenwald F, Shao B, Heinecke J, Lobos-Gonzalez L, Leyton L, Quest AFG. Inclusion of ΑVβ3 integrin into extracellular vesicles in a caveolin-1 tyrosine-14- phosphorylation dependent manner and subsequent transfer to recipient melanoma cells promotes migration, invasion and metastasis. Cell Commun Signal 2025; 23:139. [PMID: 40098186 PMCID: PMC11912626 DOI: 10.1186/s12964-025-02131-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
Caveolin-1 (CAV1) is a membrane protein that promotes migration, invasion and metastasis of cancer cells when phosphorylated on tyrosine-14 (Y14) by a cell intrinsic mechanism involving the activation of a novel Rab5-Rac1 signaling axis. Moreover, CAV1 expressed in aggressive cancer cells is included into extracellular vesicles (EVs) and such EVs increase the metastatic potential of recipient lower grade cancer cells. However, the relevance of CAV1 Y14 phosphorylation in these extrinsic EV-stimulated events remained to be determined. Here we used B16F10 mouse melanoma cells over-expressing wild-type CAV1, phospho-mimetic CAV1(Y14E) or phospho-null CAV1(Y14F) as models to determine how the EV protein content was affected by Y14 phosphorylation and how these EVs modulated the metastatic potential of recipient B16F10 cells lacking CAV1. EVs from B16F10 cells over-expressing wild-type and CAV1(Y14/E) contain CAV1, and other proteins linked to signaling pathways associated with cell adhesion and migration. CAV1 inclusion in EVs was reduced by the Y14F mutation and global protein composition was also significantly different. Moreover, CAV1 wild-type and CAV1(Y14E) EVs promoted migration, as well as invasion of cells lacking CAV1 [B16F10(Mock) cells]. In addition, β3 integrin was transferred via CAV1(Y14E) EVs to B16F10 (Mock) cells, and treatment with such EVs promoted metastasis of recipient B16F10(Mock) cells. Finally, CAV1(Y14E) EV-enhanced migration, invasion and metastasis of recipient cells was blocked by anti-αVβ3 antibodies. In conclusion, CAV1 phosphorylated on Y14 not only intrinsically promotes migration, invasion and metastasis of cells expressing the protein (in cis), but also favors the inclusion of CAV1 into EVs, as well as the extrinsic acquisition of malignant traits in recipient cells, through integrin transfer (in trans).
Collapse
Grants
- FONDECYT grants 1210644 (A.F.G.Q.), 1200836, 1240888 (L.L.), 1211223 (L.L.-G.), 1190928 (M.V.G.), FONDAP grants 15130011 and 1523A0008 (A.F.G.Q., L.L., L.L.-G., M.V.G.), ANID/BASAL/FB210008 (M.V.G.), ANID postdoctoral fellowship award Becas Chile (A.C.), 3170169 (J.D.), ANID PhD fellowship awards 21130102 (AC), 21161246 (R.H.). FONDECYT, FONDAP, ANID/BASAL/, ANID postdoctoral fellowship award Becas Chile, ANID PhD fellowship awards
Collapse
Affiliation(s)
- R Huilcaman
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile
- Facultad de Ciencias de la Salud, Escuela de Tecnología Médica, Universidad Bernardo OHiggins, General Gana 1702, Santiago, 8370854, Chile
| | - A Campos
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile
- Cancer Research UK Scotland Institute, Garscube Estate. Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | - P Contreras
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| | - L Simón
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile
- Nutrition and Dietetic School, Universidad Finis Terrae, Santiago, Chile
| | - M Varas-Godoy
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago, 7510156, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba, Santiago, 8580702, Chile
| | - F Grünenwald
- Laboratory of Reproductive Biology, Center for Biomedical Research, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Baohai Shao
- Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98195- 8055, USA
| | - Jay Heinecke
- Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98195- 8055, USA
| | - L Lobos-Gonzalez
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile
| | - L Leyton
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile.
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile.
| | - A F G Quest
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile.
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
36
|
Morabito M, Thibodot P, Gigandet A, Compagnon P, Toso C, Berishvili E, Lacotte S, Peloso A. Liver Extracellular Matrix in Colorectal Liver Metastasis. Cancers (Basel) 2025; 17:953. [PMID: 40149289 PMCID: PMC11939972 DOI: 10.3390/cancers17060953] [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: 01/30/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
The liver is the most common site of metastasis of colorectal cancer (CRC), and colorectal liver metastasis is one of the major causes of CRC-related deaths worldwide. The tumor microenvironment, particularly the extracellular matrix (ECM), plays a critical role in CRC metastasis and chemoresistance. Based on findings from clinical and basic research, this review attempts to offer a complete understanding of the role of the ECM in colorectal liver metastasis and to suggest potential ways for therapeutic intervention. First, the ECMs' role in regulating cancer cell fate is explored. We then discuss the hepatic ECM fingerprint and its influence on the metastatic behavior of CRC cells, highlighting key molecular interactions that promote metastasis. In addition, we examine how changes in the ECM within the metastatic niche contribute to chemoresistance, focusing on ECM remodeling by ECM stiffening and the activation of specific signaling pathways. Understanding these mechanisms is crucial for the development of novel strategies to overcome metastasis and improve outcomes for CRC patients.
Collapse
Affiliation(s)
- Marika Morabito
- General, Emergency and Transplant Surgery Department, ASST Settelaghi, University Hospital and Faculty of Medicine of Insubria, 21100 Varese, Italy
| | - Pauline Thibodot
- Hepato-Biliary Center, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris, 94800 Villejuif, France
| | - Anthony Gigandet
- School of Medecine, Faculty of Medecine, University of Geneva, 1211 Geneva, Switzerland
| | - Philippe Compagnon
- Division of Transplantation, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, 1205 Geneva, Switzerland;
| | - Christian Toso
- Division of Abdominal Surgery and Transplantation, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, 1205 Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, 1211 Geneva, Switzerland;
| | - Stéphanie Lacotte
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland;
| | - Andrea Peloso
- Hepato-Biliary Center, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris, 94800 Villejuif, France
- Division of Transplantation, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, 1205 Geneva, Switzerland;
- Division of Abdominal Surgery and Transplantation, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, 1205 Geneva, Switzerland
| |
Collapse
|
37
|
Youssef E, Palmer D, Fletcher B, Vaughn R. Exosomes in Precision Oncology and Beyond: From Bench to Bedside in Diagnostics and Therapeutics. Cancers (Basel) 2025; 17:940. [PMID: 40149276 PMCID: PMC11940788 DOI: 10.3390/cancers17060940] [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: 01/13/2025] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Exosomes have emerged as pivotal players in precision oncology, offering innovative solutions to longstanding challenges such as metastasis, therapeutic resistance, and immune evasion. These nanoscale extracellular vesicles facilitate intercellular communication by transferring bioactive molecules that mirror the biological state of their parent cells, positioning them as transformative tools for cancer diagnostics and therapeutics. Recent advancements in exosome engineering, artificial intelligence (AI)-driven analytics, and isolation technologies are breaking barriers in scalability, reproducibility, and clinical application. Bioengineered exosomes are being leveraged for CRISPR-Cas9 delivery, while AI models are enhancing biomarker discovery and liquid biopsy accuracy. Despite these advancements, key obstacles such as heterogeneity in exosome populations and the lack of standardized isolation protocols persist. This review synthesizes pioneering research on exosome biology, molecular engineering, and clinical translation, emphasizing their dual roles as both mediators of tumor progression and tools for intervention. It also explores emerging areas, including microbiome-exosome interactions and the integration of machine learning in exosome-based precision medicine. By bridging innovation with translational strategies, this work charts a forward-looking path for integrating exosomes into next-generation cancer care, setting it apart as a comprehensive guide to overcoming clinical and technological hurdles in this rapidly evolving field.
Collapse
|
38
|
Liu Z, Zhang X, Ben T, Li M, Jin Y, Wang T, Song Y. Focal adhesion in the tumour metastasis: from molecular mechanisms to therapeutic targets. Biomark Res 2025; 13:38. [PMID: 40045379 PMCID: PMC11884212 DOI: 10.1186/s40364-025-00745-7] [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: 11/04/2024] [Accepted: 02/11/2025] [Indexed: 03/09/2025] Open
Abstract
The tumour microenvironment is the "hotbed" of tumour cells, providing abundant extracellular support for growth and metastasis. However, the tumour microenvironment is not static and is constantly remodelled by a variety of cellular components, including tumour cells, through mechanical, biological and chemical means to promote metastasis. Focal adhesion plays an important role in cell-extracellular matrix adhesion. An in-depth exploration of the role of focal adhesion in tumour metastasis, especially their contribution at the biomechanical level, is an important direction of current research. In this review, we first summarize the assembly of focal adhesions and explore their kinetics in tumour cells. Then, we describe in detail the role of focal adhesion in various stages of tumour metastasis, especially its key functions in cell migration, invasion, and matrix remodelling. Finally, we describe the anti-tumour strategies targeting focal adhesion and the current progress in the development of some inhibitors against focal adhesion proteins. In this paper, we summarize for the first time that focal adhesion play a positive feedback role in pro-tumour metastatic matrix remodelling by summarizing the five processes of focal adhesion assembly in a multidimensional way. It is beneficial for researchers to have a deeper understanding of the role of focal adhesion in the biological behaviour of tumour metastasis and the potential of focal adhesion as a therapeutic target, providing new ideas for the prevention and treatment of metastases.
Collapse
Affiliation(s)
- Zonghao Liu
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
- The First Clinical College, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Xiaofang Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Tianru Ben
- The First Clinical College, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Mo Li
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
| | - Yi Jin
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
| | - Tianlu Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
- Department of Radiotherapy, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning Province, 110042, People's Republic of China.
- Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning Province, 116024, P. R. China.
| | - Yingqiu Song
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
- Department of Radiotherapy, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
| |
Collapse
|
39
|
Yates J, Mathey-Andrews C, Park J, Garza A, Gagné A, Hoffman S, Bi K, Titchen B, Hennessey C, Remland J, Shannon E, Camp S, Balamurali S, Cavale SK, Li Z, Raghawan AK, Kraft A, Boland G, Aguirre AJ, Sethi NS, Boeva V, Van Allen E. Cell states and neighborhoods in distinct clinical stages of primary and metastatic esophageal adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.17.608386. [PMID: 39229240 PMCID: PMC11370330 DOI: 10.1101/2024.08.17.608386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Esophageal adenocarcinoma (EAC) is a highly lethal cancer of the upper gastrointestinal tract with rising incidence in western populations. To decipher EAC disease progression and therapeutic response, we performed multiomic analyses of a cohort of primary and metastatic EAC tumors, incorporating single-nuclei transcriptomic and chromatin accessibility sequencing, along with spatial profiling. We identified tumor microenvironmental features previously described to associate with therapy response. We identified five malignant cell programs, including undifferentiated, intermediate, differentiated, epithelial-to-mesenchymal transition, and cycling programs, which were associated with differential epigenetic plasticity and clinical outcomes, and for which we inferred candidate transcription factor regulons. Furthermore, we revealed diverse spatial localizations of malignant cells expressing their associated transcriptional programs and predicted their significant interactions with microenvironmental cell types. We validated our findings in three external single-cell RNA-seq and three bulk RNA-seq studies. Altogether, our findings advance the understanding of EAC heterogeneity, disease progression, and therapeutic response.
Collapse
Affiliation(s)
- Josephine Yates
- Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland
- ETH AI Center, ETH Zurich, Zurich, Switzerland
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Camille Mathey-Andrews
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Amanda Garza
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Andréanne Gagné
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Samantha Hoffman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
| | - Kevin Bi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Breanna Titchen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
| | | | - Joshua Remland
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Erin Shannon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Sabrina Camp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Siddhi Balamurali
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Shweta Kiran Cavale
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Zhixin Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Akhouri Kishore Raghawan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Agnieszka Kraft
- Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
| | - Genevieve Boland
- Department of Surgery, Division of Gastrointestinal and Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew J Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Nilay S Sethi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Valentina Boeva
- Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland
- ETH AI Center, ETH Zurich, Zurich, Switzerland
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
- Cochin Institute, Inserm U1016, CNRS UMR 8104, Paris Descartes University UMR-S1016, Paris 75014, France
| | - Eliezer Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
- Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Fan J, Sha T, Ma B. Cancer-Derived Extracellular Vesicle ITGB2 Promotes the Progression of Triple-Negative Breast Cancer via the Activation of Cancer-Associated Fibroblasts. GLOBAL CHALLENGES (HOBOKEN, NJ) 2025; 9:2400235. [PMID: 40071217 PMCID: PMC11891567 DOI: 10.1002/gch2.202400235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/19/2024] [Indexed: 03/14/2025]
Abstract
Breast cancer is the most prevalent cancer and a leading cause of death among women globally, posing a significant public health challenge. Triple-negative breast cancer (TNBC), an aggressive subtype accounting for 15-20% of all breast cancers, lacks targeted therapies due to the absence of hormone receptors and HER2, resulting in poor prognosis and high recurrence rates. This study investigates the role of cancer-derived extracellular vesicle (EV) integrin beta-2 (ITGB2) in TNBC progression. These findings reveal that ITGB2 is significantly overexpressed in TNBC tissues and serum EVs, correlating with advanced tumor stages and poor patient survival. ITGB2 enhances TNBC progression by activating cancer-associated fibroblasts (CAFs) within the tumor microenvironment, promoting tumor growth, migration, and invasion. Mechanistic studies demonstrate that EV ITGB2 facilitates CAF activation, driving tumor-stroma interactions that support TNBC progression. These results highlight ITGB2 as a potential biomarker and therapeutic target in TNBC, emphasizing the need for novel interventions to combat this challenging breast cancer subtype.
Collapse
Affiliation(s)
- Jingjing Fan
- Xinjiang Key Laboratory of OncologyThe Affiliated Cancer Hospital of Xinjiang Medical UniversityUrumqi830011China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in XinjiangUrumqi830011China
| | - Tong Sha
- Xinjiang Key Laboratory of OncologyThe Affiliated Cancer Hospital of Xinjiang Medical UniversityUrumqi830011China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in XinjiangUrumqi830011China
| | - Binlin Ma
- Xinjiang Key Laboratory of OncologyThe Affiliated Cancer Hospital of Xinjiang Medical UniversityUrumqi830011China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in XinjiangUrumqi830011China
| |
Collapse
|
41
|
Wu S, Nasser B Singab A, Lin G, Wang Y, Zhu H, Yang G, Chen J, Li J, Li P, Zhao D, Tian J, Ye L. The regulatory role of integrin in gastric cancer tumor microenvironment and drug resistance. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:130-136. [PMID: 39798809 DOI: 10.1016/j.pbiomolbio.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/18/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
Gastric cancer (GC) remains a significant global health burden due to its high aggressiveness, early metastasis, and poor prognosis. Despite advances in chemotherapy and targeted therapies, drug resistance remains a major obstacle to improving patient outcomes. Integrins, a family of transmembrane receptors, play a pivotal role in mediating tumor growth, invasion, and drug resistance by interacting with the tumor microenvironment (TME) and regulating signaling pathways such as Wnt/β-catenin, FAK, and MAPK. This review highlights the critical functions of various integrin subunits (e.g., α5, αv, β1, β3, β6) in promoting GC progression and their involvement in chemoresistance mechanisms. Additionally, integrins modulate immune cell infiltration and stromal cell interactions within the TME, further complicating GC treatment. Emerging evidence suggests that targeting integrins, either through inhibitors or integrin-specific therapeutic strategies, holds potential in overcoming drug resistance and improving clinical outcomes. This review underscores the need for further exploration of integrins as therapeutic targets in GC and suggests promising avenues for integrin-based therapies in personalized medicine.
Collapse
Affiliation(s)
- Songlin Wu
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Abdel Nasser B Singab
- Department of Pharmacognosy, Ain-Shams University, Cairo, Egypt; Centre of Drug Discovery Research and Development, Ain Shams University, Cairo, Egypt
| | - Guimei Lin
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; National Medical Products Administration Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong University, Jinan, Shandong, China
| | - Yulu Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Huaibo Zhu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Guang Yang
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Jiaqi Chen
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Jiaxuan Li
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Peiyao Li
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Di Zhao
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Jing Tian
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Lan Ye
- Center for Cancer Prevention and Treatment, Second Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
42
|
Zhang Y, Cai Z, Zou R, Wang R, Tan R, Wang L, Wu Y, He H, He Y, Chang G. Solution-Gated Thin Film Transistor Biosensor-Based SnO 2 Amorphous Film for Label-Free Detection of Epithelial Cell Adhesion Molecules. ACS Sens 2025; 10:1187-1196. [PMID: 39888336 DOI: 10.1021/acssensors.4c03073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Epithelial cell adhesion molecule (EpCAM) was considered to be an important marker of multiple tumors, and its high expression is closely related to the early diagnosis and treatment of tumors. At present, metal oxide semiconductors have become a key component of biosensor and bioelectronics technology. Tin oxide shows great potential for development because of its nontoxic, nonpolluting, low price, and excellent electrical properties. In this study, a novel SnO2 solution-gated thin film transistor (SGTFT) biosensor for the specific detection of EpCAM was successfully developed using SnO2 film prepared by the sol-gel method as the channel material. By selecting the optimal thickness of 100 nm SnO2 film as the channel material, the transconductance value (gm) reached 1432 μS, and the threshold voltage (Vth) remained stable at 0.288 V. In order to achieve qualitative and quantitative detection of EpCAM, SnO2 films were subjected to a specific chemical treatment to fix the aptamer. Through a specific recognition between the aptamer and EpCAM, the gate voltage changes were triggered to regulate the channel current of the device. FE-SEM, EIS, XPS, and electrical performance tests were employed to track and measure the modification process. Based on the optimizations described above, the prepared SGTFT exhibited high detection sensitivity (14.6 mV·dec-1), the limit of detection (LOD) down to 24.4 pg/mL, and the calibration curves in the range of 0.02 ng/mL-500 ng/mL for EpCAM sensing. The developed SnO2-SGTFT biosensor is anticipated to provide a new highly sensitive and specific detection platform for health monitoring and disease diagnosis.
Collapse
Affiliation(s)
- Yaxing Zhang
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Zhiwei Cai
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Rong Zou
- College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| | - Ruling Wang
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Runan Tan
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Lei Wang
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Yuxiang Wu
- College of Physical Education, Jianghan University, Wuhan 430056, China
| | - Hanping He
- College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| | - Yunbin He
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Gang Chang
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| |
Collapse
|
43
|
Ludwig B, Krautkremer N, Tomassi S, Di Maro S, Di Leva FS, Benge A, Nieberler M, Kessler H, Marinelli L, Kossatz S, Reuning U. Switching Roles─Exploring Concentration-Dependent Agonistic versus Antagonistic Behavior of Integrin Ligands. J Med Chem 2025; 68:4334-4351. [PMID: 39908297 PMCID: PMC11874007 DOI: 10.1021/acs.jmedchem.4c02111] [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: 09/05/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
Identification of integrins as cancer targets has stimulated the development of specific inhibitory ligands. However, following cilengitide's unexpected clinical failure by promoting angiogenesis at low concentrations, pure ligand antagonism was soon scrutinized. We evaluated αvβ3, αvβ6, or α5β1 ligands for concentration-dependent functional switches in respective integrin subtype-overexpressing cancer cells. Cilengitide (L2) or L1 provoked minor transient changes in (p)-FAK and (p)-p44/42(erk-1/2) predominantly at low concentrations and antagonized cell migration at high concentrations, while agonistically accelerating it at low concentrations. L5 (α5β1) showed bell-shaped FAK activation at both concentrations, blocking cell migration at high concentrations only in α5β1+ OV-MZ-6 cells, not acting agonistically. L3 (αvβ6) did not alter signaling upon long exposure but transiently and early activated FAK in αvβ6+ HN cells at both concentrations, with neither antagonistic nor agonistic consequences on cell motility. These data underscore the need for in-depth evaluation of ligand actions to ensure their most promising medical use.
Collapse
Affiliation(s)
- Beatrice
Stefanie Ludwig
- Department
of Nuclear Medicine, School of Medicine & Health, Klinikum rechts
der Isar, TUM University Hospital, Technical
University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
- Central Institute
for Translational Cancer Research (TranslaTUM), School of Medicine
& Health, Klinikum rechts der Isar, TUM University Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
| | - Nils Krautkremer
- Department
of Oral and Maxillofacial Surgery, School of Medicine & Health,
Klinikum rechts der Isar, TUM University
Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
| | - Stefano Tomassi
- UNINA
−
Department of Pharmacy, University of Naples
Federico II, Via Domenico Montesano 49, Naples 80131, Italy
| | - Salvatore Di Maro
- SUN −
Department of Environmental, Biological and Pharmaceutical Sciences
and Technologies, Università degli
Studi della Campania “Luigi Vanvitelli”, Viale Abramo Lincoln, 5, Caserta 81100, Italy
| | - Francesco Saverio Di Leva
- UNINA
−
Department of Pharmacy, University of Naples
Federico II, Via Domenico Montesano 49, Naples 80131, Italy
| | - Anke Benge
- Department
of Obstetrics & Gynecology, School of Medicine & Health, Clinical
Research Unit, Klinikum rechts der Isar, TUM University Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
| | - Markus Nieberler
- Department
of Oral and Maxillofacial Surgery, School of Medicine & Health,
Klinikum rechts der Isar, TUM University
Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
| | - Horst Kessler
- Department
of Chemistry, School of Natural Sciences and Bavarian NMR Center (BNMRZ), Institute for Advanced Study, Technical University
Munich, Lichtenbergstrasse
2a, Garching 85748, Germany
| | - Luciana Marinelli
- UNINA
−
Department of Pharmacy, University of Naples
Federico II, Via Domenico Montesano 49, Naples 80131, Italy
| | - Susanne Kossatz
- Department
of Nuclear Medicine, School of Medicine & Health, Klinikum rechts
der Isar, TUM University Hospital, Technical
University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
- Central Institute
for Translational Cancer Research (TranslaTUM), School of Medicine
& Health, Klinikum rechts der Isar, TUM University Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
- Department
of Chemistry, School of Natural Sciences, Technical University Munich, Ismaninger Strasse 22, Munich 81675, Germany
| | - Ute Reuning
- Department
of Obstetrics & Gynecology, School of Medicine & Health, Clinical
Research Unit, Klinikum rechts der Isar, TUM University Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
| |
Collapse
|
44
|
Zhou S, Li B, Wu D, Chen Y, Zeng W, Huang J, Tan L, Mao G, Liu F. Mechanisms of fibrinogen trans-activation of the EGFR/Ca2+ signaling axis to regulate mitochondrial transport and energy transfer and inhibit axonal regeneration following cerebral ischemia. J Neuropathol Exp Neurol 2025; 84:210-222. [PMID: 39495964 DOI: 10.1093/jnen/nlae114] [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] [Indexed: 11/06/2024] Open
Abstract
Ischemic stroke results in inhibition of axonal regeneration but the roles of fibrinogen (Fg) in neuronal signaling and energy crises in experimental stroke are under-investigated. We explored the mechanism of Fg modulation of axonal regeneration and neuronal energy crisis after cerebral ischemia using a permanent middle cerebral artery occlusion (MCAO) rat model and primary cortical neurons under low glucose-low oxygen. Behavioral tests assessed neurological deficits; immunofluorescence, immunohistochemistry, and Western-blot analyzed Fg and protein levels. Fluo-3/AM fluorescence measured free Ca2+ and ATP levels were gauged via specific assays and F560nm/F510nm ratio calculations. Mito-Tracker Green labeled mitochondria and immunoprecipitation studied protein interactions. Our comprehensive study revealed that Fg inhibited axonal regeneration post-MCAO as indicated by reduced GAP43 expression along with elevated free Ca2+, both suggesting an energy crisis. Fg impeded mitochondrial function and mediated impairment through the EGFR/Ca2+ axis by trans-activating EGFR via integrin αvβ3 interaction. These results indicate that the binding of Fg with integrin αvβ3 leads to the trans-activation of the EGFR/Ca2+ signaling axis thereby disrupting mitochondrial energy transport and axonal regeneration and exacerbating the detrimental effects of ischemic neuronal injury.
Collapse
Affiliation(s)
- Shengqiang Zhou
- National TCM Master Liu Zuyi Inheritance Studio, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha City, Hunan Province, China
| | - Bo Li
- Department of Pediatrics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha City, Hunan Province, China
| | - Dahua Wu
- Department of Neurology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha City, Hunan Province, China
| | - Yanjun Chen
- Graduate School, Hunan University of Chinese Medicine, Changsha City, Hunan Province, China
| | - Wen Zeng
- Graduate School, Hunan University of Chinese Medicine, Changsha City, Hunan Province, China
| | - Jia Huang
- Graduate School, Hunan University of Chinese Medicine, Changsha City, Hunan Province, China
| | - Lingjuan Tan
- Graduate School, Hunan University of Chinese Medicine, Changsha City, Hunan Province, China
| | - Guo Mao
- Key Project Office, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha City, Hunan Province, China
| | - Fang Liu
- National TCM Master Liu Zuyi Inheritance Studio, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha City, Hunan Province, China
| |
Collapse
|
45
|
Okuyama T, Tsuno T, Inoue R, Fukushima S, Kyohara M, Matsumura A, Miyashita D, Nishiyama K, Takano Y, Togashi Y, Meguro-Horike M, Horike SI, Kin T, Shapiro AJ, Yanagisawa H, Terauchi Y, Shirakawa J. The matricellular protein Fibulin-5 regulates β-cell proliferation in an autocrine/paracrine manner. iScience 2025; 28:111856. [PMID: 39995864 PMCID: PMC11848788 DOI: 10.1016/j.isci.2025.111856] [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: 02/22/2024] [Revised: 11/20/2024] [Accepted: 01/17/2025] [Indexed: 02/26/2025] Open
Abstract
The matricellular protein Fibulin-5 (Fbln5) is a secreted protein that is essential for elastic fiber formation, and pancreatic islets are usually surrounded by the extracellular matrix (ECM), which includes elastic fibers. However, much uncertainty remains regarding the function of the ECM and its components in β-cells. Here, we describe the role of Fbln5 in β-cell replication. Fbln5 expression was increased upon glucose stimulation in β-cells of mouse and human islets. β-Cell-specific Fbln5-knockout (βFbln5KO) mice exhibit significantly reduced β-cell proliferation in vivo but not in vitro. Secreted extracellular Fbln5 enhances β-cell replication. Fbln5-deficient β-cells exhibit the downregulated expression of the gene encoding Polo-like kinase 1 (PLK1), which is accompanied by ERK-mediated FoxM1 nuclear export. These data suggest that Fbln5 is secreted from β-cells in response to glucose and plays important roles in the appropriate maintenance of β-cell functions in an autocrine or paracrine manner.
Collapse
Affiliation(s)
- Tomoko Okuyama
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Takahiro Tsuno
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
| | - Ryota Inoue
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
| | - Setsuko Fukushima
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
| | - Mayu Kyohara
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Anzu Matsumura
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
| | - Daisuke Miyashita
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Kuniyuki Nishiyama
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
| | - Yusuke Takano
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Yu Togashi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Makiko Meguro-Horike
- Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Shin-ichi Horike
- Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Tatsuya Kin
- Clinical Islet Laboratory and Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - A.M. James Shapiro
- Clinical Islet Laboratory and Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Jun Shirakawa
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
| |
Collapse
|
46
|
Baster Z, Russell L, Rajfur Z. A Review of Talin- and Integrin-Dependent Molecular Mechanisms in Cancer Invasion and Metastasis. Int J Mol Sci 2025; 26:1798. [PMID: 40076426 PMCID: PMC11899650 DOI: 10.3390/ijms26051798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer is the second most common cause of death in the world, representing one of the main economic burdens in health care and research. The effort of research has mainly focused on limiting the growth of a localized tumor, but most recently, there has been more attention focused on restricting the spreading of the cancer via invasion and metastasis. The signaling pathways behind these two processes share many molecules with physiological pathways regulating cell adhesion and migration, and, moreover, adhesion and migration processes themselves underlie tumor potential for invasion. In this work, we reviewed the latest literature about cancer development and invasion and their regulation by cell migration- and adhesion-related proteins, with a specific focus on talins and integrins. We also summarized the most recent developments and approaches to anti-cancer therapies, concentrating on cell migration-related therapies.
Collapse
Affiliation(s)
- Zbigniew Baster
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland
- Laboratory for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lindsay Russell
- Undergraduate Program, Barnard College of Columbia University, New York, NY 10027, USA;
| | - Zenon Rajfur
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland
- Jagiellonian Center of Biomedical Imaging, Jagiellonian University, 30-348 Kraków, Poland
| |
Collapse
|
47
|
Evangelista A, Scocozza F, Conti M, Auricchio F, Conti B, Dorati R, Genta I, Benazzo M, Pisani S. Exploring Mechanical Features of 3D Head and Neck Cancer Models. J Funct Biomater 2025; 16:74. [PMID: 40137353 PMCID: PMC11942903 DOI: 10.3390/jfb16030074] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) presents significant challenges in oncology due to its complex biology and poor prognosis. Traditional two-dimensional (2D) cell culture models cannot replicate the intricate tumor microenvironment, limiting their usefulness in studying disease mechanisms and testing therapies. In contrast, three-dimensional (3D) in vitro models provide more realistic platforms that better mimic the architecture, mechanical features, and cellular interactions of HNSCC. This review explores the mechanical properties of 3D in vitro models developed for HNSCC research. It highlights key 3D culture techniques, such as spheroids, organoids, and bioprinted tissues, emphasizing their ability to simulate critical tumor characteristics like hypoxia, drug resistance, and metastasis. Particular attention is given to stiffness, elasticity, and dynamic behavior, highlighting how these models emulate native tumor tissues. By enhancing the physiological relevance of in vitro studies, 3D models offer significant potential to revolutionize HNSCC research and facilitate the development of effective, personalized therapeutic strategies. This review bridges the gap between preclinical and clinical applications by summarizing the mechanical properties of 3D models and providing guidance for developing systems that replicate both biological and mechanical characteristics of tumor tissues, advancing innovation in cancer research and therapy.
Collapse
Affiliation(s)
- Aleksandra Evangelista
- Department of Otorhinolaryngology, Fondazione IRCCS Policlinico San Matteo, Via Golgi 19, 27100 Pavia, Italy; (A.E.); (M.B.)
| | - Franca Scocozza
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy; (M.C.); (F.A.)
| | - Michele Conti
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy; (M.C.); (F.A.)
- 3D and Computer Simulation Laboratory, IRCCS Policlinico San Donato, Piazza Edmondo Malan 2, San Donato Milanese, 20097 Milano, Italy
| | - Ferdinando Auricchio
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy; (M.C.); (F.A.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (B.C.); (R.D.); (I.G.); (S.P.)
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (B.C.); (R.D.); (I.G.); (S.P.)
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (B.C.); (R.D.); (I.G.); (S.P.)
| | - Marco Benazzo
- Department of Otorhinolaryngology, Fondazione IRCCS Policlinico San Matteo, Via Golgi 19, 27100 Pavia, Italy; (A.E.); (M.B.)
| | - Silvia Pisani
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (B.C.); (R.D.); (I.G.); (S.P.)
| |
Collapse
|
48
|
Qin Q, Feng M, Zhang K, Mo Z, Liu Y, Ma Y, Liu X. Basigin in cerebrovascular diseases: Roles, mechanisms, and therapeutic target potential. Eur J Pharmacol 2025; 989:177232. [PMID: 39734038 DOI: 10.1016/j.ejphar.2024.177232] [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: 09/22/2024] [Revised: 11/24/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Cerebrovascular diseases are major global health issues, responsible for significant morbidity and mortality. Basigin (additionally called CD147 or EMMPRIN) is a glycosylated transmembrane protein that facilitates intercellular communication. Recent research has highlighted the critical role of Basigin in inducing matrix metalloproteinases (MMPs), which contribute to the progression of cerebrovascular diseases. Consequently, Basigin has emerged as a promising therapeutic target for these conditions. However, inhibiting the pivotal role of Basigin in mediating cerebrovascular disease is an urgent area of investigation. In this review, we systematically examine the pathological mechanisms by which Basigin contributes to the development of cerebrovascular diseases. We present evidence demonstrating the protective effect of targeted inhibition of Basigin in these conditions and suggest future research directions.
Collapse
Affiliation(s)
- Qi Qin
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou City, Henan Province, 450000, China
| | - Mengzhao Feng
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou City, Henan Province, 450000, China
| | - Kaiyuan Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou City, Henan Province, 450000, China
| | - Zhizhun Mo
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1 Fuhua Road, Shenzhen City, Guangdong Province, 518033, China
| | - Yuxiang Liu
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1 Fuhua Road, Shenzhen City, Guangdong Province, 518033, China
| | - Yinzhong Ma
- Institute of Medicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Nanshan District, Shenzhen City, Guangdong Province, 518055, China.
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou City, Henan Province, 450000, China.
| |
Collapse
|
49
|
Featherby SJ, Faulkner EC, Maraveyas A, Ettelaie C. Identification of the Interacting Domains Between Tissue Factor and β1-Integrin and the Signalling Properties of the Two Fibronectin-like Domains of Tissue Factor. Cancers (Basel) 2025; 17:644. [PMID: 40002240 PMCID: PMC11853675 DOI: 10.3390/cancers17040644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Interactions between tissue factor (TF) and β1-integrin induce cell signals, but the molecular mechanisms are not completely understood. The extracellular domain of TF and EGF4-βTD domains of β1-integrin were hypothesised to be the most likely domains involved in the interaction. Additionally, the interaction may induce a conformational change in β1-integrin, which results in changes in signalling. METHODS Peptide constructs corresponding to the upper (residues 1-110; UED), lower (residues 106-219; LED) or combined extracellular domain (residues 1-219; TED) of TF were produced, as well as peptides corresponding to EGF4-βTD or EGF4 domains of β1-integrin. These constructs were expressed in TF-rich MDA-MB-231 cells and TF-deficient primary endothelial cells. The association of the peptides with endogenous-TF or β1-integrin was assessed by a proximity ligation assay and co-immunoprecipitation. Additionally, the influence of the constructs on β1-integrin conformation and the outcome on ERK1/2 activation, cyclin D expression and cell proliferation was analysed. RESULTS In MDA-MB-231 cells, all TF-constructs were associated with β1-integrin whilst LED was co-immunopurified with β1-integrin. EGF4-βTD was associated with and co-immunopurified with endogenous TF. Additionally, the expression of UED or EGF4-βTD reduced ERK phosphorylation and cyclin D expression and suppressed proliferation. In endothelial cells, the expression of UED, and to a lesser extent, LED, reduced the proportion of β1-integrin in the active conformation and induced ERK1/2 phosphorylation but did not induce cyclin D expression or proliferation. CONCLUSIONS Collectively, these data indicate the extracellular domains of TF function together, with the lower domain forming a robust interaction with the βTD of β1-integrin and the upper domain inducing cell signalling by regulating β1-integrin conformation.
Collapse
Affiliation(s)
- Sophie J. Featherby
- Centre for Biomedicine, Hull-York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK; (E.C.F.); (C.E.)
| | - Eamon C. Faulkner
- Centre for Biomedicine, Hull-York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK; (E.C.F.); (C.E.)
| | - Anthony Maraveyas
- Clinical Sciences, Hull-York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK;
| | - Camille Ettelaie
- Centre for Biomedicine, Hull-York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK; (E.C.F.); (C.E.)
| |
Collapse
|
50
|
Shi X, Zang J, Gu Q, Zhang M, Sun H, Yang L, Cheng J, Wang R, Mao H, Xu A, Wang X, Xiao Y, Cai J, Han F, Yang D, Li Y, Nie H. Comprehensive analysis of the multifaceted role of ITGAV in digestive system cancer progression and immune infiltration. Front Immunol 2025; 16:1480771. [PMID: 40018050 PMCID: PMC11864929 DOI: 10.3389/fimmu.2025.1480771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/24/2025] [Indexed: 03/01/2025] Open
Abstract
Background Digestive system cancers are among the most common malignancies, exhibiting consistently high incidence and mortality rates, yet effective detection and treatment targets remain limited. Integrin αv (ITGAV, CD51) is a significant member of the integrin family, widely recognized for its role in mediating interactions between cells and the extracellular matrix, as well as intracellular signaling. In recent years, ITGAV has been found to have significantly elevated expression in multiple tumors, such as prostate cancer, breast cancer, and osteosarcoma, and was considered to be a key component in various stages of tumor progression. However, no systematic digestive system cancer analysis has been conducted to explore its function in prognosis, diagnosis, and immunology. Methods Transcriptome sequencing and clinical data of samples were obtained from The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression Project (GTEx), Human Protein Atlas (HPA), cBioPortal, TIMER and TISIDB databases. Bioinformatics methods were employed to investigate the potential oncogenicity of ITGAV, focusing specifically on the analysis of its prognosis, diagnostic value, and immune infiltration level of ITGAV in digestive system cancers. In addition, GO, KEGG, and PPI network analysis revealed the biological functions and related signaling pathways related to ITGAV. Finally, the role of ITGAV in regulating cancer progression was experimentally verified using hepatocellular carcinoma and pancreatic cancer as examples. Results We found that ITGAV was highly expressed in multiple digestive system cancers. In addition, high expression of ITGAV was closely associated with poor prognosis and showed potential for early diagnosis. Enrichment of pathways related to extracellular matrix organizing processes and tumor migratory movements was identified. In vitro, results showed that the knockdown of ITGAV significantly inhibited the migratory movement ability of hepatocellular carcinoma and pancreatic cancer cells, while its overexpression significantly promoted the migration of the above cells. Finally, immunoassays showed a significant correlation between ITGAV expression and the infiltration level of various immune cells, further clarifying the critical role of ITGAV in the tumor immune microenvironment. Conclusion Our results elucidated the importance of ITGAV in the prognostic assessment, early diagnosis, and targeted immunotherapy of digestive system cancers, and revealed its multifaceted role in regulating cancer progression.
Collapse
Affiliation(s)
- Xinyue Shi
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Jingyu Zang
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Qi Gu
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Mengmeng Zhang
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Handi Sun
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Lijun Yang
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Jiahui Cheng
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Ruonan Wang
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Han Mao
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Aitong Xu
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Xin Wang
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Yu Xiao
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Jialing Cai
- Department of Toxicology, College of Public Health, Harbin Medical University, Harbin, Heilongjiang, China
| | - Fang Han
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Depeng Yang
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Yu Li
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Huan Nie
- School of Life Science and Technology, Faculty of Life Sciences and Medicine, Harbin Institute of Technology, Harbin, Heilongjiang, China
| |
Collapse
|