101
|
Araujo-Castro M, Álvarez-Escola C, Casteràs A, Carmona-Bayonas A, Chiara MD, Hanzu FA, Hernando J, Vercher-Conejero JL, Rodríguez-Fraile M, Gómez Dos Santos V, Jimenez-Fonseca P, Giraldo A, Valdés N, Vidal O, Del Olmo-García M, Capdevila J. Spanish consensus on the diagnosis and management of adrenocortical carcinoma. Endocr Relat Cancer 2025; 32:e250034. [PMID: 40215284 PMCID: PMC12053981 DOI: 10.1530/erc-25-0034] [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: 02/11/2025] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy with an estimated incidence of 0.7-2 cases per million/year. The rarity of this disease, coupled with limited preclinical models and clinical trials, has hindered progress, resulting in poor outcomes, with a 5-year survival rate of approximately 35%. Currently, the only available curative treatment is complete surgical resection of the adrenal tumor. For unresectable or metastatic ACC, the current standard therapeutic modalities are mitotane, chemotherapy, radiotherapy and locoregional treatments; however, these are noncurative. Mitotane has an adrenolytic and anti-steroidogenic effect, and it is used in the adjuvant setting for high-risk patients, as systemic therapy for metastatic disease, and/or to control hormonal secretion. While key pathways in ACC pathogenesis have been identified as potential therapeutic targets, results with targeted therapies remain modest, showing that there is a clinical unmet need for novel treatments or new combinations of exiting drugs. Effective management requires a multidisciplinary team of experts to optimize outcomes for patients. This article presents a multidisciplinary consensus on the diagnosis, management, prognosis and follow-up of patients with ACC, and the approach to two special contexts, ACC in pregnant women and hormone-producing ACC. The consensus was coordinated by the Spanish Society of Endocrinology and Nutrition (SEEN) and the Spanish Group of Neuroendocrine and Endocrine Tumors (GETNE), with contribution from experts from related societies including the Spanish Association of Surgeons (AEC), Spanish Society of Urology (AEU), Anatomic-Pathology (SEAP), Nuclear Medicine (SEMNIM), Medical Oncology (SEOM) and Radiotherapeutic Oncology (SEOR).
Collapse
Affiliation(s)
- Marta Araujo-Castro
- Endocrinology & Nutrition Department. Hospital Universitario Ramón y Cajal Madrid, Madrid, Spain
- Instituto de Investigación Biomédica Ramón y Cajal (IRYCIS), Madrid, Spain
| | | | - Ana Casteràs
- Endocrinology & Nutrition Department. Hospital Universitario Vall d´Hebron, Barcelona, Spain
| | - Alberto Carmona-Bayonas
- Medical Oncology Department, Hospital General Universitario Morales Meseguer, University of Murcia, IMIB, Murcia, Spain
| | - María-Dolores Chiara
- Health Research Institute of the Principality of Asturias, Oviedo, Spain
- Institute of Oncology of the Principality of Asturias, University of Oviedo, Oviedo, Spain
| | - Felicia A Hanzu
- Endocrinology & Nutrition Department, Hospital Clinic, University of Barcelona, IDIBPAS, CIBERDEM, Barcelona, Spain
| | - Jorge Hernando
- Medical Oncology Department, Gastrointestinal and Endocrine Tumor Unit, Hospital Universitario Vall D'Hebron, Vall Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | | | - Victoria Gómez Dos Santos
- Instituto de Investigación Biomédica Ramón y Cajal (IRYCIS), Madrid, Spain
- Urology Department, Hospital Universitario Ramón y Cajal Madrid, Madrid, Spain
| | - Paula Jimenez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Alexandra Giraldo
- Radiotherapy Oncology Department, Hospital Universitario Vall D' Hebron, VHIO, Barcelona, Spain
| | - Nuria Valdés
- Endocrinology & Nutrition Department, Hospital Universitario Cruces, Barakaldo, Spain
- UPV/EHU, Biobizkaia, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Oscar Vidal
- General Surgery Department. Hospital Clinic de Barcelona, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Maribel Del Olmo-García
- Endocrinology & Nutrition Department. Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Jaume Capdevila
- Medical Oncology Department, Gastrointestinal and Endocrine Tumor Unit, Hospital Universitario Vall D'Hebron, Vall Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
102
|
Ebadi AH, Moghimian M, Abtahi-Evari SH, Saadatian Z, Fathi A, Soltani M. Chrysin Modulates Behavior and Hippocampal Histopathology in Adult Male Hypoandrogenic Rats: The Regulatory Role of miR-30a, miR-375, and miR-204. Brain Behav 2025; 15:e70529. [PMID: 40329827 DOI: 10.1002/brb3.70529] [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: 11/26/2024] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
OBJECTIVE Hypoandrogenism, a condition linked to oxidative stress and hippocampal cell death, can lead to learning and memory disorders in aging men or those with hypogonadism. Recently, microRNAs such as miR-30a, miR-375, and miR-204 have been identified as regulators of cell death and memory formation. Flavonoid antioxidants that modulate microRNA expression offer a potential therapeutic approach for neurodegenerative conditions. This study examines the effects of the flavonoid chrysin on these microRNAs, cell death processes, and memory under hypoandrogenic stress. METHODS Rats were assigned to six groups: control, hypoandrogenic, hypoandrogenic treated with chrysin (50 mg/kg and 75 mg/kg), and chrysin treated alone (50 mg/kg and 75 mg/kg). After 14 days, memory changes were assessed using the Morris water maze test, along with evaluations of oxidative stress enzymes, apoptosis and autophagy, miR-30a, miR-375, and miR-204 gene expression, and the number of dark neurons. RESULTS Hypoandrogenism increased apoptosis and autophagy gene expression, dark neurons, oxidative stress enzymes, and decreased memory and microRNA expression compared to controls. Treatment with chrysin (75 mg/kg) significantly reduced apoptosis and autophagy gene expression, dark neurons, and oxidative stress enzymes, and improved memory and learning. This group also showed modulation of miR-30a, miR-375, and miR-204 levels compared to the hypoandrogenic group. CONCLUSION Hypoandrogenism led to neurodegeneration and memory impairment, likely due to oxidative stress affecting miR-30a, miR-375, and miR-204 regulation. Chrysin (75 mg/kg) potentially mitigates cell death processes and memory deficits in hypoandrogenism by modulating these microRNAs.
Collapse
Affiliation(s)
- Amir-Hossein Ebadi
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Maryam Moghimian
- Department of Physiology, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Seyd-Hossein Abtahi-Evari
- Department of Biochemistry and Nutrition, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Zahra Saadatian
- Department of Physiology, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Alireza Fathi
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Malihe Soltani
- Department of Anatomy, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
103
|
Wong V, Goodstein T, Montenegro GB, Srinivasan R, Singer EA. Biomarkers in advanced renal cell carcinoma: current practice and future directions. Curr Opin Oncol 2025; 37:274-282. [PMID: 40156235 PMCID: PMC11970984 DOI: 10.1097/cco.0000000000001138] [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: 04/01/2025]
Abstract
PURPOSE OF REVIEW This review focuses on contemporary research into potential prognostic and therapeutic biomarkers for advanced renal cell carcinoma (RCC) published over the past 18 months. Beyond serum lab values, there is no consensus on the use of specific biomarkers for this purpose. Potential biomarkers being investigated consist of genetic, protein, immunologic, and radiologic candidates. RECENT FINDINGS New insights in genomic biomarkers include a better understanding of VHL mutational heterogeneity, tumor mutational burden, and the importance of genes like PBRM1 and SETD2 . Protein biomarkers such as C-reactive protein (CRP) and PDZK1 have demonstrated utility in predicting disease progression, therapeutic response, and survival, while immunologic biomarkers like PSMD2, cytokines, and Tregs continue to shed light on the tumor microenvironment and immune evasion. Emerging imaging biomarkers, from CAIX-targeted radiotracers to PSMA-based PET-CT, offer noninvasive diagnostic and prognostic tools that may revolutionize RCC management. SUMMARY There are several promising biomarkers currently under investigation for use in advanced RCC.
Collapse
Affiliation(s)
- Vivian Wong
- Division of Urologic Oncology, The Ohio State University James Comprehensive Cancer Center, Columbus, OH
| | | | - Gabriela Bravo Montenegro
- Molecular Therapeutics Section, Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ramaprasad Srinivasan
- Molecular Therapeutics Section, Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Eric A. Singer
- Division of Urologic Oncology, The Ohio State University James Comprehensive Cancer Center, Columbus, OH
| |
Collapse
|
104
|
Chanpanitkitchote P, Nuanpirom J, Pongsapich W, Asavapanumas N, Mendler S, Wiesmann N, Brieger J, Jinawath N. EMILIN-1 Suppresses Cell Proliferation through Altered Cell Cycle Regulation in Head and Neck Squamous Cell Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:995-1012. [PMID: 39892781 DOI: 10.1016/j.ajpath.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/24/2024] [Accepted: 01/15/2025] [Indexed: 02/04/2025]
Abstract
Extracellular matrix (ECM) proteins play an important role in the pathological processes of tumor development and progression. Elastic microfibril interface located protein-1 (EMILIN-1), an ECM glycoprotein, is linked to cell adhesion and migration. It was identified from head and neck squamous cell carcinoma (HNSCC) tissues that down-regulated EMILIN-1. It is associated with an increased risk of secondary primary malignancy development in HNSCC and hypothesized to function as a tumor suppressor in HNSCC. This study showed that EMILIN-1 expression in HNSCC tissues was specific to the stromal area, and secreted-EMILIN-1 level was higher in fibroblasts isolated from HNSCC tissues than in HNSCC cells. EMILIN-1 overexpression decreased cell proliferation, migration, and invasion in FaDu and CAL27 cells. Knockdown of EMILIN-1 in HNSCC cancer-associated fibroblasts induced cell proliferation and migration. The conditioned medium from EMILIN-1 knockdown cancer-associated fibroblasts increased HNSCC cell proliferation, and the co-culture system enhanced cancer cell migration and invasion. RNA-sequencing analysis revealed that the cell cycle and aurora kinase signaling were the most significant enrichment pathways, confirmed at the protein level. Furthermore, in an in ovo chick chorioallantoic membrane model, overexpression of EMILIN-1 in FaDu cells reduced tumor size and Ki-67-positivity and increased cleaved caspase-3-positive cells. These findings suggest that EMILIN-1 suppresses HNSCC growth partly through the down-regulation of cell cycle and aurora kinase signaling pathways.
Collapse
Affiliation(s)
| | - Jiratchaya Nuanpirom
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand; Integrative Computational BioScience Center (ICBS), Mahidol University, Nakhon Pathom, Thailand
| | - Warut Pongsapich
- Department of Otorhinolaryngology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nithi Asavapanumas
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Simone Mendler
- Department of Otorhinolaryngology, University Medical Center Mainz, Mainz, Germany
| | - Nadine Wiesmann
- Department of Otorhinolaryngology, University Medical Center Mainz, Mainz, Germany; Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Mainz, Germany
| | - Juergen Brieger
- Department of Otorhinolaryngology, University Medical Center Mainz, Mainz, Germany
| | - Natini Jinawath
- Integrative Computational BioScience Center (ICBS), Mahidol University, Nakhon Pathom, Thailand; Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand; Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
105
|
Asmat-Abanto AS, Portocarrero-Reyes A, Espejo-Carrera RE, Minchón-Medina CA, Timaná-Palacios DJ. Bibliometric analysis in Scopus of scientific production on the relationship between periodontitis and gastrointestinal cancer (2014 - 2023). Med Oral Patol Oral Cir Bucal 2025; 30:e422-e430. [PMID: 39864084 PMCID: PMC12019650 DOI: 10.4317/medoral.26969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/07/2025] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND Recent studies have suggested that some opportunistic periodontal pathobionts have oncogenic properties. However, few bibliometric studies investigate the relationship between periodontitis and gastrointestinal cancer. This bibliometric study aimed to analyze these epidemiological studies conducted between 2014 and 2023 to guide future research. MATERIAL AND METHODS In March 2024, the Scopus database was explored. The articles selected were subjected to a bibliometric analysis of study designs, trends in annual scientific production, and networks of collaboration among countries. Furthermore, the most outstanding countries, academic institutions, authors and journals with the most significant number of publications and the top most cited articles were analyzed. For this purpose, Microsoft Excel, SPSS and VOSviewer 1.6.20 were used. RESULTS A total of 123 documents were included for analysis. China contributed the most significant number of publications (33 articles) and the United States had the most significant number of citations (2709). Weimin Ye (h-index:81) and Dominique Michaud (h-index:73) were the most prolific authors (5 articles); Dominique Michaud also had the most citations (470). International Journal of Cancer was the journal with the highest number of articles published (6), in addition to being the publication that had the most citations of these articles (409). Tufts University in the United States had the highest number of citations. CONCLUSIONS Between 2014 and 2023, 123 articles on the relationship between periodontitis and gastrointestinal cancer were published. The largest scientific production was found in China, and the most cited articles were those from the United States. Likewise, the research design most commonly used was the cross-sectional type.
Collapse
Affiliation(s)
- A-S Asmat-Abanto
- 3145 America Sur Avenue Monserrate Neighborhood, Trujillo 13008, Peru
| | | | | | | | | |
Collapse
|
106
|
Bushi A, Ma Y, Adu-Amankwaah J, Wang R, Cui F, Xiao R, Zhao J, Yuan J, Tan R. G protein-coupled estrogen receptor biased signaling in health and disease. Pharmacol Ther 2025; 269:108822. [PMID: 39978643 DOI: 10.1016/j.pharmthera.2025.108822] [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: 08/30/2024] [Revised: 02/03/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
G protein-coupled estrogen receptor (GPER) is now recognized for its pivotal role in cellular signaling, influencing diverse physiological processes and disease states. Unlike classical estrogen receptors, GPER exhibits biased signaling, wherein ligand binding triggers selective pathways over others, significantly impacting cellular responses. This review explores the nuanced mechanisms of biased signaling mediated by GPER, underscoring its relevance in cardiovascular health, neurological function, immune modulation, and oncogenic processes. Despite its critical implications, biased signaling through GPER remains underexplored compared to traditional signaling paradigms. We explore recent progress in understanding GPER signaling specificity and its potential therapeutic implications across various diseases. Future research directions aim to uncover the molecular basis of biased signaling, develop selective ligands, and translate these insights into personalized therapeutic approaches. Exploiting the therapeutic potential of GPER biased signaling represents a promising frontier in precision medicine, offering innovative strategies to address unmet medical needs.
Collapse
Affiliation(s)
- Aisha Bushi
- School international education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yixuan Ma
- First Clinical Medical School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Joseph Adu-Amankwaah
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Rong Wang
- The second clinical college, China Medical University, Shenyang, Liaoning 110122, China
| | - Fen Cui
- Research Institution of Behavioral Medicine Education, Jining Medical University, Jining 272067, China
| | - Rui Xiao
- Second Clinical Medical School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jinming Zhao
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China; Department of Pathology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Jinxiang Yuan
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong, China.
| | - Rubin Tan
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
107
|
Yu# W, Shi# Y, Bao X, Chen X, Ni Y, Wang J, Ye H. Hyaluronan-mediated motility receptor-mediated aerobic glycolysis enhances stem-like properties and chemoresistance in lung adenocarcinoma. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2025; 29:337-347. [PMID: 40051130 PMCID: PMC12012315 DOI: 10.4196/kjpp.24.275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 04/22/2025]
Abstract
Lung adenocarcinoma (LUAD) is a global malignancy with significant chemoresistance impacting patient prognosis. The pro-tumorigenic role of hyaluronan- mediated motility receptor (HMMR) in LUAD is recognized. This study was designed to investigate the underlying mechanisms by which HMMR affects chemoresistance in LUAD. Bioinformatics presented the expression patterns of HMMR in LUAD patients and the association between HMMR levels and patient survival, followed by qRT-PCR to verify HMMR expression in LUAD tissues and cells. Further, bioinformatics was leveraged to identify the signaling pathways enriched by HMMR and its relevance to glycolytic genes, we also analyzed changes in the glycolytic activity of LUAD cells by manipulating HMMR expression. Stemness was evaluated through cell aggregation assays and Western blot, and drug responsiveness was gauged using CCK-8 assays, alongside flow cytometry for apoptosis analysis. HMMR was highly expressed in LUAD tissues and cells, and this overexpression correlated with poorer prognoses in patients. GSEA showed that HMMR was notably enriched in the glycolysis and gluconeogenesis pathways, correlating positively with the expression of key glycolytic genes. Cellular experiments confirmed that HMMR knockdown notably suppressed aerobic glycolysis in LUAD cells. Moreover, overexpression of HMMR could further enhance the stemness and cisplatin resistance of LUAD cells by stimulating glycolysis. In brief, this study has validated that high levels of HMMR in LUAD are predictive of poor patient prognosis, and that overexpression of HMMR can catalyze aerobic glycolysis, thus promoting stemness and chemoresistance in LUAD cells. Thus, HMMR could be a target for improving chemosensitivity in LUAD.
Collapse
Affiliation(s)
- Wenwen Yu#
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing 325600, China
| | - Yubo Shi#
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing 325600, China
| | - Xiaoqiong Bao
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing 325600, China
| | - Xiangxiang Chen
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing 325600, China
| | - Yangyang Ni
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing 325600, China
| | - Jincong Wang
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing 325600, China
| | - Hua Ye
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing 325600, China
| |
Collapse
|
108
|
Shao Y, Duan B, Li H, Li X, Peng S, Zheng H, You Z. Target Screening and Single Cell Analysis of Diabetic Retinopathy and Hepatocarcinoma. J Cell Mol Med 2025; 29:e70521. [PMID: 40293350 DOI: 10.1111/jcmm.70521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 04/30/2025] Open
Abstract
The association between liver cancer and diabetes has been a longstanding focus in medical research. Current evidence suggests that diabetes is an independent risk factor for the development of liver cancer. Diabetic retinopathy (DR), a prevalent neurovascular complication of diabetes, has yet to be fully characterised concerning liver cancer. Therefore, this study seeks to identify shared genes and pathways between liver cancer and DR to uncover potential therapeutic targets. Immune infiltration and cell communication in liver cancer were analysed using the GEO single-cell dataset GSM7494113. Single-cell RNA sequencing data from rat retinas were obtained from the GEO datasets GSE209872 and GSE160306. Ferritin phagocytosis-related genes were retrieved from the GeneCards database. The SeuratR package was employed for single-cell clustering analysis, while the CellChat package assessed differences in intercellular communication. Genes shared between DR and liver cancer were identified, and the DGIDB database was consulted to predict potential drug-gene interactions targeting membrane proteins involved in ferritin phagocytosis. Key ferritin phagocytosis (FRHG) genes were further validated using quantitative real-time polymerase chain reaction (qRT-PCR). After annotating the single-cell data through dimensionality reduction and clustering, the expression of genes associated with membrane protein-related ferritinophagy was notably elevated in both HCC and DR samples. Based on the expression of ferritinophagy-related genes, the ferritin deposition score in Müller cells from the DR group was significantly higher than that in the control group. Cell communication analysis revealed that central hub genes associated with ferritinophagy, such as PSAP and MK, along with other signalling pathways, were significantly upregulated in the high Müller group compared to the low Müller group. In contrast, VEGF expression was enhanced in the low Müller group. Importantly, the machine learning model constructed using these key hub genes demonstrated high diagnostic efficacy for both HCC and DR. Finally, by simulating a hyperosmotic diabetic microenvironment, we confirmed in vitro that high glucose conditions significantly stimulate the expression of the shared key hub genes in both HCC and DR. The present study identified the connection between ferritinophagy-related subgroups of cells and key hub genes in both HCC and DR, providing new insights into DR-associated biomarkers and the shared pathological regulatory pathways with HCC. These findings further suggest potential therapeutic targets for both diseases.
Collapse
Affiliation(s)
- Yinan Shao
- School of Optometry, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Institute of Ophthalmology and Vision Science, Nanchang, China
- Key Laboratory of Ophthalmology of Jiangxi Province, Nanchang, China
- Jiangxi Branch Center, National Clinical Research Center for Eye Diseases, Nanchang, China
- Affiliated Eye Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Clinical Research Center for Eye Diseases, Nanchang, China
| | - Bingfen Duan
- School of Optometry, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Institute of Ophthalmology and Vision Science, Nanchang, China
- Key Laboratory of Ophthalmology of Jiangxi Province, Nanchang, China
- Jiangxi Branch Center, National Clinical Research Center for Eye Diseases, Nanchang, China
- Affiliated Eye Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Clinical Research Center for Eye Diseases, Nanchang, China
| | - Haotian Li
- The First Affiliated Hospital of Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Xiaonan Li
- School of Optometry, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Institute of Ophthalmology and Vision Science, Nanchang, China
- Key Laboratory of Ophthalmology of Jiangxi Province, Nanchang, China
- Jiangxi Branch Center, National Clinical Research Center for Eye Diseases, Nanchang, China
- Affiliated Eye Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Clinical Research Center for Eye Diseases, Nanchang, China
| | - Shijing Peng
- School of Optometry, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Institute of Ophthalmology and Vision Science, Nanchang, China
- Key Laboratory of Ophthalmology of Jiangxi Province, Nanchang, China
- Jiangxi Branch Center, National Clinical Research Center for Eye Diseases, Nanchang, China
- Affiliated Eye Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Clinical Research Center for Eye Diseases, Nanchang, China
| | - Haowen Zheng
- School of Optometry, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Institute of Ophthalmology and Vision Science, Nanchang, China
- Key Laboratory of Ophthalmology of Jiangxi Province, Nanchang, China
- Jiangxi Branch Center, National Clinical Research Center for Eye Diseases, Nanchang, China
- Affiliated Eye Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Clinical Research Center for Eye Diseases, Nanchang, China
| | - Zhipeng You
- School of Optometry, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Institute of Ophthalmology and Vision Science, Nanchang, China
- Key Laboratory of Ophthalmology of Jiangxi Province, Nanchang, China
- Jiangxi Branch Center, National Clinical Research Center for Eye Diseases, Nanchang, China
- Affiliated Eye Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Clinical Research Center for Eye Diseases, Nanchang, China
| |
Collapse
|
109
|
Aromokeye R, Ackerman-Berrier M, Araujo RDC, Lambousis M, Cardoza S, Chen L, Kaplan ME, Zhu H, Zerbinatti C, Penton C, Thatcher GRJ, Marlowe T. Development of a high-throughput TR-FRET assay to identify inhibitors of the FAK-paxillin protein-protein interaction. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025:100237. [PMID: 40318755 DOI: 10.1016/j.slasd.2025.100237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/21/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Focal Adhesion Kinase (FAK) is a non-receptor tyrosine kinase and scaffolding protein that is primarily regulated by integrin signaling. FAK signaling increases cell motility in both normal and cancer cells, and FAK is often overexpressed and/or dysregulated in many types of cancer. FAK has three different domains: an N-terminal FERM domain, a central kinase domain (the traditional target for drug discovery), and a C-terminal focal adhesion targeting (FAT) domain. The FAT domain represents an alternative approach to targeting FAK, and our aim is to identify novel small molecules that will inhibit FAT protein-protein interactions (PPI), which may have implications for cancer and fibrosis treatment. Here, we describe the development and validation of a robust high-throughput screening (HTS) assay suitable for identifying inhibitors of the FAT:paxillin PPI. The 384-well low volume assay is based on time-resolved fluorescence resonance energy transfer (TR-FRET) technology and uses the high affinity biotin-PEG-1907 stapled peptide to mimic paxillin. We also present the development of a TR-FRET counterscreen assay using CD47 and SIRPα to detect nonspecific inhibitors, as well as an orthogonal surface plasmon resonance (SPR) binding assay. We employed the FAT: biotin-PEG-1907 assay to screen a 31,636-compound small molecule library. Primary positives (hits) from HTS were confirmed in concentration-response primary and counterscreen assays and validated in the SPR binding assay. We discovered 4 inhibitors of the FAT:paxillin PPI using this approach and established a framework for small molecule drug discovery efforts targeting the FAT domain of FAK.
Collapse
Affiliation(s)
- Rukayat Aromokeye
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Martha Ackerman-Berrier
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Rosa Del Carmen Araujo
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Maria Lambousis
- University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Savio Cardoza
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Lie Chen
- Arizona Center for Drug Discovery, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Matthew E Kaplan
- Functional Genomics Core, Research, Innovation and Impact, University of Arizona, Tucson, AZ, USA
| | - Haining Zhu
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Celina Zerbinatti
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA; Arizona Center for Drug Discovery, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Christopher Penton
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA; Arizona Center for Drug Discovery, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Gregory R J Thatcher
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA.
| | - Timothy Marlowe
- University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA.
| |
Collapse
|
110
|
Guo S, Ding R, Zhao Q, Wang X, Lv S, Ji XY. Recent Insights into the Roles of PEST-Containing Nuclear Protein. Mol Biotechnol 2025; 67:1800-1813. [PMID: 38762838 DOI: 10.1007/s12033-024-01188-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 04/26/2024] [Indexed: 05/20/2024]
Abstract
PEST-containing nuclear protein (PCNP), a short-lived small nuclear protein with 178 amino acids, is a nuclear protein containing two PEST sequences. PCNP is highly expressed in several malignant tumors such as cervical cancer, rectal cancer, and lung cancer. It is also associated with cell cycle regulation and the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) and Wnt signaling pathways during tumor growth. The present article discuss how PCNP regulates the PI3K/AKT/mTOR and Wnt signaling pathways and related proteins, and the ubiquitination of PCNP regulates tumor cell cycle as well as the progress of the application of PCNP in the pathophysiology and treatment of colon cancer, human ovarian cancer, thyroid cancer, lung adenocarcinoma and oral squamous cell carcinoma. The main relevant articles were retrieved from PubMed, with keywords such as PEST-containing nuclear protein (PCNP), cancer (tumor), and signaling pathways as inclusion/exclusion criteria. Relevant references has been included and cited in the manuscript.
Collapse
Affiliation(s)
- Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Ruidong Ding
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Qian Zhao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Xu Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Shuangyu Lv
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
- Kaifeng Key Laboratory for Infectious Diseases and Biosafety, Kaifeng, 475004, Henan, China.
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Mazhai, Erqi District, Zhengzhou, 450064, Henan, China.
| |
Collapse
|
111
|
Song J, Kang F, Chang X, Wu J, Zhang P, Wang X, Wang F. Irradiation of 125I seeds blocks glycolysis in pancreatic cancer by inhibiting KLF5 m6A methylation through the suppression of RBM15. Exp Cell Res 2025; 448:114538. [PMID: 40147709 DOI: 10.1016/j.yexcr.2025.114538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 03/29/2025]
Abstract
This paper investigated whether 125I seed irradiation for pancreatic cancer treatment was mediated through the RBM15/KLF5 pathway in glycolysis. The study collected peripheral blood from pancreatic cancer patients, and detected the expression of RBM15 and KLF5 expression in the serum of pancreatic cancer patients before and after 125I seed irradiation. An in vitro study was conducted to investigate the effects 125I seed irradiation on the malignant behavior and glycolysis of pancreatic cancer cells. The underlying mechanisms were thoroughly examined through a series of logical experiments, including Western blot analysis, Dot-blot experiment, methylated RNA immunoprecipitation assay, and RNA pull down assay. A xenograft tumor model in nude mice was established and treated with 125I seed irradiation, which was employed to research the in vivo effect and mechanism of 125I seed irradiation for pancreatic cancer. The overexpressed RBM15 and KLF5 in serum of pancreatic cancer patients were reduced after 125I seed treatment. 125I seed treatment impaired pancreatic cancer cell proliferation and invasion; enhanced apoptosis; attenuated glycolysis; and reduced RBM15 and KLF5 expression. RBM15 overexpression partially reversed these influences of 125I seed treatment on pancreatic cancer cells. RBM15 was capable of increasing KLF5 expression, which might be achieved by promoting m6A methylation of KLF5. In vivo, 125I seed treatment blocked the growth of pancreatic cancer cells and decreased RBM15 and KLF5 expression in xenograft tumor, whereas RBM15 overexpression abolished these effects. 125I seed irradiation suppressed glycolysis in pancreatic cancer by inhibiting KLF5 m6A methylation through down-regulation of RBM15. This discovery established a solid foundation for the use of in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Jianjun Song
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, China
| | - Fuping Kang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, China
| | - Xiaoning Chang
- Department of Rehabilitation Medicine, General Hospital of Ningxia Medical University, China
| | - Jing Wu
- Medical Experiment Center, General Hospital of Ningxia Medical University, China
| | - Peng Zhang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, China
| | - Xiaolong Wang
- School of Nursing, Zhengzhou Institute of Industrial Application Technology, China
| | - Feng Wang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, China.
| |
Collapse
|
112
|
Zhang W, Wang S, Xing Y, Luo X, Wang R, Yu F. Bioorthogonal SERS-bioluminescence dual-modal imaging for real-time tracking of triple-negative breast cancer metastasis. Acta Biomater 2025; 197:431-443. [PMID: 40101869 DOI: 10.1016/j.actbio.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025]
Abstract
Triple-negative breast cancer (TNBC) represents an aggressive subtype of breast cancer, characterized by early metastasis and a poor prognosis. Traditional imaging modalities often lack the sensitivity and molecular specificity required for the early detection of metastatic lesions. In this study, we developed a dual-modal imaging strategy that integrates surface-enhanced Raman scattering (SERS) and bioluminescence imaging probes, utilizing bioorthogonal labeling to track TNBC organ metastasis. The SERS probes were encapsulated with azide-labeled macrophage membranes to extend circulation time and enhance targeting efficiency. Additionally, bioorthogonal metabolic glycolengineering was employed to modify luciferase-labeled tumor cells (4T1-Luc) with bicyclo[6.1.0]nonyne (BCN) groups, facilitating precise binding between the probes and 4T1-Luc cells through click chemistry reactions. This dual-modal imaging approach enabled real-time monitoring of small metastatic lesions with high sensitivity, providing a non-invasive and accurate method for assessing tumor metastasis and therapeutic response in vivo. Our findings indicate that the dual-modal imaging technique, combining SERS and bioluminescence with bioorthogonal labeling, holds significant potential for advanced applications in oncology. STATEMENT OF SIGNIFICANCE: This study devised a surface-enhanced Raman scattering (SERS) and bioluminescence dual-modal imaging strategy integrated with a bioorthogonal label to address the challenge of tracking the metastasis of aggressive triple-negative breast cancer (TNBC). In contrast to conventional methods, this approach facilitated real-time, whole-body monitoring of tumor dissemination through bioluminescence. Simultaneously, it achieved the detection of micro-metastases in organs using SERS, thereby exceeding the sensitivity limitations of existing imaging techniques. Clinical validation with human samples further demonstrated its potential for non-invasive therapeutic assessment and early intervention. By bridging preclinical innovation and clinical requirements, this research offered a transformative tool for precision oncology. It is expected to attract the interest of researchers in the fields of biomedicine, nanotechnology, and cancer therapeutics.
Collapse
Affiliation(s)
- Wei Zhang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China
| | - Sisi Wang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China; Department of Breast and Thyroid Surgery, The Second Affiliated Hospital, Hainan Medical University, Haikou 571199, PR China
| | - Yanlong Xing
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China
| | - Xianzhu Luo
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China
| | - Rui Wang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China.
| | - Fabiao Yu
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China.
| |
Collapse
|
113
|
Tang Z, Zhang Z, Zhao J, Zhang F, Zhang Y, Wen Y, Li M, Sun J, Shi L, Chen W, Li Z, Guo Z, Liu Y. Integrated analysis of multiple programmed cell death-related prognostic genes and functional validation of apoptosis-related genes in osteosarcoma. Int J Biol Macromol 2025; 307:142113. [PMID: 40089239 DOI: 10.1016/j.ijbiomac.2025.142113] [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: 07/04/2024] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
Osteosarcoma (OS) is one of the most prevalent bone malignancies with a poor prognosis. Various types of programmed cell death patterns can influence cancer progression and response to treatment. We aimed to integrate different molecular characteristics of cell death for risk stratification and personalized therapy. First, we obtained transcriptomic, single-cell transcriptomic, and clinical information from the TARGET-OS and GEO databases as well as analyzed genes in fourteen cell death patterns to establish the cell death index (CDI) signature. A nomogram constructed from the CDI calculated from seven genes in combination with metastasis could effectively predict the prognosis of OS patients. Subsequently, the prognostic value and immune characteristics in CDI-defined subgroups were analyzed. A construct nomogram model was also constructed with clinical information. Notably, immunohistochemistry confirmed that the expression of GALNT14, a core gene in CDI model, correlated with poor survival. Deficiency of the highly expressed prognostic gene GALNT14 significantly repressed OS progression and OS cell proliferation by promoting apoptosis. We subsequently demonstrated that Bortezomib, a targeted inhibitor of GALNT14, can be used to enhance chemosensitivity. Finally, it was further elucidated that Bortezomib reduces MT2A glycosylation and improves its stability to promote apoptosis in OS cells by inhibiting GALNT14 expression. In summary, integration of multiple cell death genes may improve the ability to stratify risk in patients with OS, and targeting GALNT14 with Bortezomib improves chemotherapy sensitivity and induces apoptosis.
Collapse
Affiliation(s)
- Zhen Tang
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China; Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Zhi Zhang
- Department of Orthopedic Surgery, Guyuan People's Hospital, Ningxia, China
| | - Jungang Zhao
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Fan Zhang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Yiran Zhang
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Yanhua Wen
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Miaozhen Li
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jin Sun
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Lei Shi
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Wentian Chen
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Zheng Li
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China.
| | - Zheng Guo
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China.
| | - Yingxiang Liu
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China.
| |
Collapse
|
114
|
Fassnacht M, Puglisi S, Kimpel O, Terzolo M. Adrenocortical carcinoma: a practical guide for clinicians. Lancet Diabetes Endocrinol 2025; 13:438-452. [PMID: 40086465 DOI: 10.1016/s2213-8587(24)00378-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 03/16/2025]
Abstract
Adrenocortical carcinoma is a rare endocrine malignancy. The management of patients with adrenocortical carcinoma is challenging for several reasons, including its heterogeneous but frequently aggressive biological behaviour; tumour-related hormonal excess (eg, Cushing's syndrome or virilisation); the overall paucity of evidence regarding diagnostic investigation and treatment; the approval of only one drug (mitotane); and the scarcity of centres with sufficient experience. In this Review, we present 25 questions on the most important aspects of the clinical management of adult patients with adrenocortical carcinoma that we have frequently asked ourselves over the past 25 years. We offer our personal answers and perspectives, drawing upon published evidence as well as more than 60 years of collective clinical experience and insights from our management of more than 1700 patients across two centres in Germany and Italy.
Collapse
Affiliation(s)
- Martin Fassnacht
- Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany; Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany; National Center for Tumor Diseases WERA, Würzburg, Germany.
| | - Soraya Puglisi
- Department of Clinical and Biological Sciences, Internal Medicine, San Luigi Hospital, University of Turin, Turin, Italy
| | - Otilia Kimpel
- Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Massimo Terzolo
- Department of Clinical and Biological Sciences, Internal Medicine, San Luigi Hospital, University of Turin, Turin, Italy
| |
Collapse
|
115
|
Huang L, Ye B, Cao F, Ruan B, Li X. Single-Cell Atlas of the Peripheral Immune Response in Patients With Chronic Hepatitis B. J Med Virol 2025; 97:e70360. [PMID: 40255189 DOI: 10.1002/jmv.70360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/22/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025]
Abstract
Cellular immune responses are crucial in determining outcomes of the hepatitis B virus (HBV) infection. Ineffective immune responses enable persistent HBV infection and contribute to progressive liver disease. Understanding the mechanisms underlying immunological HBV tolerance and restoring functional adaptive immune responses is essential for successful chronic hepatitis B (CHB) treatment. This study examined the dysregulated immune responses and immunopathological cell states associated with CHB using single-cell RNA sequencing of peripheral blood mononuclear cells to investigate immune cell composition and transcriptional differences between patients with CHB and healthy donors. Phenotypic alterations in the lymphoid and myeloid compartments were observed following HBV infection. T cell immune profiling in patients with CHB showed enrichment of exhausted CD8+ T cells, impaired cytotoxicity of effector CD8+ T cells, and increased regulatory T cell (Treg) suppressive activity. Immature neutrophils and a unique CD14+ monocyte subset (myeloid-derived suppressor cells) exhibited potent immunosuppressive abilities. A novel population of CD14+CD163+VSIG4+ M2-like macrophages with immunosuppressive and anti-inflammatory phenotypes was enriched in a patient with severe CHB and liver failure, indicating a potential contribution to dysfunctional immune responses. Our study demonstrated immune exhaustion and evasion in chronic HBV infection, elucidating its immunopathological features and suggesting new therapeutic strategies for immune-mediated disorders and unresolved chronic HBV infection.
Collapse
Affiliation(s)
- Li Huang
- Zhejiang Key Laboratory of Clinical In Vitro Diagnostic Techniques, Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bo Ye
- Zhejiang Key Laboratory of Clinical In Vitro Diagnostic Techniques, Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Feinan Cao
- Zhejiang Key Laboratory of Clinical In Vitro Diagnostic Techniques, Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bing Ruan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xuefen Li
- Zhejiang Key Laboratory of Clinical In Vitro Diagnostic Techniques, Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
116
|
Ekaterina K, Saba I, Sophia A, Ivane A, Maia Z. Evaluating the Diagnostic Potential of Biomarker Panels in Breast Cancer and Prostate Adenocarcinoma. Health Sci Rep 2025; 8:e70796. [PMID: 40309623 PMCID: PMC12040734 DOI: 10.1002/hsr2.70796] [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: 12/23/2024] [Revised: 03/27/2025] [Accepted: 04/16/2025] [Indexed: 05/02/2025] Open
Abstract
Background Noninvasive diagnostic methods are essential for early cancer detection and improved patient outcomes. Circulating biomarkers, measurable indicators of pathological processes, offer a promising avenue, yet optimal panels for reliable cancer diagnosis remain undefined. This study evaluates the diagnostic performance of selected plasma biomarkers in distinguishing breast cancer and prostate adenocarcinoma patients from healthy individuals, using statistical analysis and machine learning. Materials and Methods We analyzed blood samples from 162 participants (73 cancer patients: 51 with breast cancer and 22 with prostate adenocarcinoma; 89 healthy controls). Levels of 12 cancer-associated biomarkers-including Ki67, DNMT1, BRCA1, and MPO-were quantified using enzyme-linked immunosorbent assays (ELISA). Statistical analyses, including the Mann-Whitney U test and machine learning models (random forest), were employed to assess the predictive accuracy of these biomarkers in distinguishing between cancerous and healthy states. Results Biomarkers such as Ki67, DNMT1, and MPO were significantly elevated in cancer groups. Random forest models using selected combinations (e.g., BRCA1-CTA-TP53) achieved perfect classification accuracy (AUC = 1.00). However, high inter-marker correlations suggested potential redundancy, underscoring the need for biomarker panel optimization. Conclusion Our findings support the potential of biomarker panels for accurate, noninvasive cancer diagnostics. Further validation in larger, more diverse cohorts is warranted to establish clinical utility and generalizability.
Collapse
Affiliation(s)
| | - Iordanishvili Saba
- Petre Shotadze Tbilisi Medical AcademyTbilisiGeorgia
- Institute of Medical and Public Health ResearchIlia State UniversityTbilisiGeorgia
| | - Adamia Sophia
- Institute of Medical and Public Health ResearchIlia State UniversityTbilisiGeorgia
| | - Abiatari Ivane
- Institute of Medical and Public Health ResearchIlia State UniversityTbilisiGeorgia
| | - Zarnadze Maia
- Petre Shotadze Tbilisi Medical AcademyTbilisiGeorgia
| |
Collapse
|
117
|
Yang D, Yang C, Huang L, Guan M, Song C. Role of ubiquitination-driven metabolisms in oncogenesis and cancer therapy. Semin Cancer Biol 2025; 110:17-35. [PMID: 39929409 DOI: 10.1016/j.semcancer.2025.02.004] [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: 08/26/2024] [Revised: 01/17/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025]
Abstract
Ubiquitination represents one of the most critical post-translational modifications, comprising a multi-stage enzyme process that plays a pivotal role in a myriad of cellular biological activities. The deregulation of the processes of ubiquitination and deubiquitination is associated with the development of cancers and other diseases. This typescript reviews the impact of ubiquitination on metabolic processes, elucidating the regulatory functions of ubiquitination on pivotal enzymes within metabolic pathways in pathological contexts. It underscores the role of ubiquitination-driven metabolism disorders in the etiology of cancers, and oncogenesis, and highlights the potential therapeutic efficacy of targeting ubiquitination-driven enzymes in cancer metabolism, their combination with immune checkpoint inhibitors, and their clinical applications.
Collapse
Affiliation(s)
- Dongqin Yang
- Department of Laboratory Medicine of Huashan Hospital, Fudan University, Shanghai 200040, China; Central Laboratory, Huashan Hospital, Fudan University, 12 Middle Urumuqi Road, Shanghai 200040, China
| | - Can Yang
- Department of Laboratory Medicine of Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Linlin Huang
- Central Laboratory, Huashan Hospital, Fudan University, 12 Middle Urumuqi Road, Shanghai 200040, China
| | - Ming Guan
- Department of Laboratory Medicine of Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Chunhua Song
- Division of Hematology, The Ohio State University Wexner Medical Center, the James Cancer Hospital, Columbus, OH 43210, USA.
| |
Collapse
|
118
|
Zhang W, Jia X, Lou D, Wu Q, Yan Y, Mao F. Comparison of mTOR inhibitors combined with endocrine therapy versus that alone in breast cancer: a meta-analysis. Future Oncol 2025; 21:1417-1427. [PMID: 40152674 PMCID: PMC12051556 DOI: 10.1080/14796694.2025.2485022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND This meta-analysis aims to evaluate the efficacy and safety of rapamycin (mTOR) inhibitors with endocrine therapy versus endocrine therapy alone in treating advanced or metastatic estrogen receptor/progesterone receptor (ER/PR) + breast cancer. METHODS We conducted a comprehensive search in PubMed, Web of Science, Embase, and the Cochrane Library for randomized controlled trials (RCTs) comparing mTOR inhibitors plus endocrine therapy with endocrine therapy alone up to September 2024. RESULTS This analysis included 10 RCTs comprising 3,337 patients. Relative to endocrine therapy alone, the combination of mTOR inhibitors and endocrine therapy significantly improved the clinical benefit rate (RR = 1.41, p < 0.001), overall response rate (RR = 1.40, p = 0.006), progression-free survival (PFS; HR = 0.67, p < 0.001), and overall survival (OS; HR = 0.86, p = 0.056), although the improvement in OS was not statistically significant. Subgroup analyses indicated a more pronounced PFS advantage in patients under 65 years of age (HR = 0.55, p = 0.013) and those who had previously received chemotherapy (HR = 0.51, p = 0.001). However, the incidence of adverse events was higher in the combination therapy group, notably stomatitis (p < 0.001), elevated aspartate aminotransferase/alanine aminotransferase (p = 0.04), and diarrhea (p = 0.01). CONCLUSIONS The combination of mTOR inhibitors with endocrine therapy offers superior efficacy with manageable toxicities in patients with advanced or metastatic ER/PR+ breast cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Xinru Jia
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Dandi Lou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Qingping Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yici Yan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Feiyan Mao
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
119
|
Saadh MJ, Ahmed HH, Kareem RA, Bishoyi AK, Roopashree R, Shit D, Arya R, Joshi KK, Sameer HN, Yaseen A, Athab ZH, Adil M. The hidden messengers: Tumor microenvironment-derived exosomal ceRNAs in gastric cancer progression. Pathol Res Pract 2025; 269:155905. [PMID: 40073646 DOI: 10.1016/j.prp.2025.155905] [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: 11/26/2024] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in the development and progression of gastric cancer (GC). The TME comprises a network of cancer cells, immune cells, fibroblasts, endothelial cells, and extracellular matrix components, which provide a supportive niche for cancer cells. This study investigates the role of TME-derived exosomal competitive endogenous RNAs (ceRNAs), particularly long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), as major regulating agents in GC development. Exosomal ceRNAs control gene expression across several TME components, amplifying cancer hallmarks like cell proliferation, invasion, metastases, and chemoresistance. They promote dynamic interplay between cancer cells and adjacent stromal cells, enabling tumor development through immune suppression, angiogenesis, and epithelial-mesenchymal transition (EMT). Exosomal ceRNAs can modify the TME, creating a pro-tumorigenic milieu and preparing cancer cells to avoid immunological responses, defy death, and adapt to therapeutic pressures. This review highlights the understudied interactions between the TME and exosomal ceRNAs in gastric cancer and emphasizes their potential utility as diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | | | | | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Debasish Shit
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, Uttarakhand 248002, India; Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | |
Collapse
|
120
|
Yokota M, Morikawa A, Matsuoka H, Nagahisa Y, Kawata K, Okabe M, Masui T, Kawamoto K. Minimally Invasive Robotic Surgery in Lateral Lymph Node Dissection by Pelvic Floor First Approach. Dis Colon Rectum 2025; 68:e200-e201. [PMID: 39907247 DOI: 10.1097/dcr.0000000000003507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Affiliation(s)
- Mitsuru Yokota
- Department of General Surgery, Kurashiki Central Hospital, Kurashiki, Okayama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
121
|
Abu-Lafi S, Falah M, Zeidan M, Rayan M, Rayan A. Expanding paclitaxel's therapeutic window: Investigating the pharmacokinetic, clinical formulation and mechanistic aspects of paclitaxel‑lipoate conjugate. Oncol Lett 2025; 29:216. [PMID: 40093869 PMCID: PMC11907689 DOI: 10.3892/ol.2025.14962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 08/14/2024] [Indexed: 03/19/2025] Open
Abstract
Paclitaxel (PTX) is among the most widely used antimicrotubular chemotherapy agents available from natural sources. It has a wide range of antitumor effectiveness, particularly against breast, ovarian and lung malignancies. IDD-1040 is a novel anticancer chemical conjugate that combines lipoic acid with PTX and demonstrates an anticancer efficiency superior to that of PTX alone. The aim of the present study was to investigate the analytical, formulation and pharmacokinetic aspects of IDD-1040, shedding light on its pharmacological behavior and the possible mechanisms underlying its enhanced anticancer activity. IDD-1040 was administered to mice as an intravenous bolus, and the pharmacokinetic parameters were determined over the following 7 days. The results revealed a total clearance of 1.689 l/h.kg, volume of distribution of 1.93 l/kg, average half-life of 1.14 h and terminal half-life of 8.64 h. Notably, the area under the curve of IDD-1040 was >14-fold higher than that of PTX, suggesting slower metabolism and that prodrug itself may have antitumor activity. An in vitro tubulin polymerization assay revealed distinct tubulin-binding characteristics for IDD-1040 compared with PTX. Due to the poor water solubility of IDD-1040, a formulation development experiment was conducted. In total, 31 formulations were prepared that became transparent when diluted with water. In addition, some formulations achieved a relatively high drug content (12 mg/g) without the use of surfactants. Moreover, they included fewer excipients compared with the formulations diluted with water, suggesting a promising approach for drug formulation. In summary, IDD-1040 exhibited extended circulation, efficient tissue distribution and reduced metabolite formation in vitro, warranting further exploration of its mechanisms of action and therapeutic potential. Future studies are recommended to assess the stability, pharmacokinetics and pharmacodynamics of these refined IDD-1040 formulations to gauge their suitability for clinical application.
Collapse
Affiliation(s)
- Saleh Abu-Lafi
- Faculty of Pharmacy, Al-Quds University, Abu-Deis 144, Palestine
| | - Mizied Falah
- Research Center, Holy Family Hospital, Nazareth 1600000, Israel
| | - Mahmoud Zeidan
- Molecular Genetics and Virology Laboratory, Qasemi Research Center, Faculty of Science, Al-Qasemi Academic College, Baka El-Garbiah 3010000, Israel
| | - Mouhammad Rayan
- Geriatric Department, Holy Family Hospital, Nazareth 16000, Israel
| | - Anwar Rayan
- Department of Science and Technology, Faculty of Science, Al-Qasemi Academic College, Baka El-Garbiah 3010000, Israel
| |
Collapse
|
122
|
Jia H, Bian Y, Yuan J, Zhang Y, Zhang S. The Potential Role of C4 MYH11+ Fibroblasts and the MDK-SDC2 Ligand-Receptor Pair in Lung Adenocarcinoma: Implications for Prognosis and Therapeutic Strategies. Transl Oncol 2025; 55:102364. [PMID: 40121996 PMCID: PMC11982484 DOI: 10.1016/j.tranon.2025.102364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) posed a significant threat to global human health. This study employed single-cell RNA sequencing (scRNA-seq) to analyze transcriptomic data from nine LUAD patients at different stages of tumor infiltration, aiming to elucidate the tumor microenvironment and key biological processes of LUAD. METHODS In this study, we processed the scRNA-seq data using the Seurat package and sequentially applied principal component analysis followed by the Harmony package to effectively correct for batch effects, identifying 105,725 high-quality cells. Through cell clustering and gene expression profiling, we identified critical cell subpopulations and gene expression patterns in LUAD patients. RESULTS Our analysis revealed that the C4 MYH11+ Fibroblasts subtype was primarily involved in biological processes related to muscle function. Further investigations uncovered the MDK-SDC2 ligand-receptor pair as a critical regulator of tumor cell invasion, proliferation, and migration, driving LUAD progression. Additionally, we developed a gene-based prognostic model that effectively predicted patient survival, providing valuable clinical insights. CONCLUSION This study provided a comprehensive atlas of the LUAD tumor microenvironment, highlighted the role of the C4 MYH11+ Fibroblasts in tumor progression. It also proposed the MDK-SDC2 ligand-receptor pair as a novel mechanism, addressing a significant gap in this area of research. And presented a gene-based prognostic model as a novel perspective for research into immunotherapy and drug sensitivity in LUAD.
Collapse
Affiliation(s)
- Hongling Jia
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.; The first clinical medical college of Shandong university of Traditional Chinese Medicine, Jinan, China
| | - Yanjie Bian
- Xinxiang Medical University, Xinxiang, China
| | - Jie Yuan
- Sijing Town Community Healthcare Center, Shanghai, China
| | - Yi Zhang
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China..
| | - Shengyi Zhang
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China..
| |
Collapse
|
123
|
Mui J, Sarofim M, Cheng E, Gilmore A. Laparoscopic natural orifice specimen extraction for diverticular disease: a systematic review. Surg Endosc 2025; 39:3049-3056. [PMID: 40140081 PMCID: PMC12041039 DOI: 10.1007/s00464-025-11683-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Diverticular disease is extremely common in the Western world, with a proportion of complications requiring colonic resection. Whilst laparoscopic surgery has its benefits, a large wound for specimen extraction predisposes to surgical site infection, prolonged pain and prolonged hospital admission. Natural orifice specimen extraction (NOSE) is an alternative technique that has not yet been widely adopted in diverticular disease surgery. The aim of this systematic review is to evaluate the evidence on the outcomes of NOSE in left sided resections for diverticular disease. METHODS A systematic review of PubMed, Ovid MEDLINE and EMBASE was performed to identify studies that reported outcomes for left sided resections with NOSE in diverticular disease. The studies reviewed were all human studies published in a peer-reviewed journal after 2010. The participants had to be over the age of 18 and the extraction site had to be transanal/transrectal. Articles that were not full text or not in English were excluded. These studies were assessed independently by two reviewers using a standardised pre-piloted form. RESULTS One hundred and eighty-seven articles were screened, with 9 articles meeting the inclusion criteria. The study sample size ranged from 8 to 157 participants, with a pooled total of 428 patients who had NOSE. Hospital length of stay varied from 4 to 6 days. Only 2 patients required conversion to transabdominal extraction. Pain scores were lower post-NOSE compared to traditional abdominal extraction in 2 out of 3 studies. The anastomotic leak rate varied from 0 to 18%. Six studies reported no surgical site infections and there was only 1 mortality. CONCLUSION NOSE is a safe and feasible option for patients requiring left sided resection for diverticular disease based on the current available data. The literature demonstrates low rates of surgical site infection, mortality and reduced hospital length of stay.
Collapse
Affiliation(s)
- Jasmine Mui
- Department of Colorectal Surgery, Liverpool Hospital, Liverpool, NSW, Australia.
- St George and Sutherland Clinical School, University of New South Wales, Kogarah, NSW, Australia.
| | - Mina Sarofim
- Department of Colorectal Surgery, Liverpool Hospital, Liverpool, NSW, Australia
- St George and Sutherland Clinical School, University of New South Wales, Kogarah, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Ernest Cheng
- Department of Colorectal Surgery, Liverpool Hospital, Liverpool, NSW, Australia
- St George and Sutherland Clinical School, University of New South Wales, Kogarah, NSW, Australia
| | - Andrew Gilmore
- Department of Colorectal Surgery, Liverpool Hospital, Liverpool, NSW, Australia
- School of Medicine, Western Sydney University, Liverpool, NSW, Australia
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie Park, NSW, Australia
| |
Collapse
|
124
|
Deng Y, Zhang X. Multidimensional predictive model of survival risk in small cell lung cancer patients: A comprehensive analysis based on clinical characteristics and treatment strategies. Curr Probl Surg 2025; 66:101759. [PMID: 40306865 DOI: 10.1016/j.cpsurg.2025.101759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 05/02/2025]
Affiliation(s)
- Yi Deng
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Zhang
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
125
|
Chang Z, Chen B, Wang S, Chen K, Huang L, Yang Y, Wu H, Jian W, Cheng ZJ, Han X, Sun B. Organ-specific cancer biomarker identification: a ten-year single-center study in southern China. BMC Cancer 2025; 25:820. [PMID: 40312330 PMCID: PMC12044899 DOI: 10.1186/s12885-025-14225-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Cancer biomarker discovery is essential for early detection and monitoring, yet there is a lack of comprehensive studies examining organ-specific biomarkers across various cancer types. In this study, we analyzed clinical data from 59,184 cancer patients diagnosed between 2013 and 2023, focusing on 11 major cancer systems. We used propensity score matching with 55,010 healthy controls to create balanced comparison groups. Serum biomarker profiles were assessed through principal component analysis, differential expression analysis, and ROC curve analysis. Our findings revealed organ-specific biomarker patterns, such as decreased CA724, ferritin, and β2-microglobulin in thoracic cancer, reduced serum phosphorus in neurological cancer, and elevated cystatin C and creatinine in urinary system cancer. Further analysis across 22 cancer types uncovered additional biomarkers, including elevated ALT in hepatobiliary cancer, altered coagulation factors in laryngeal cancer, increased monocytes in pancreatic cancer, and reduced complement C3 in intestinal cancer. These results provide valuable insights into the unique biomarker signatures for different cancers, contributing to the potential development of more targeted and efficient screening methods.
Collapse
Affiliation(s)
- Zhenglin Chang
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, XingDaoHuanBei Road, Guangzhou, 510005, Guangdong Province, China
| | - Bingsen Chen
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Suilin Wang
- Department of Orthopedics, Guangzhou Orthopedic Hospital, Guangzhou, 510045, Guangdong Province, China
| | - Kaipai Chen
- Department of Stomatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Linliang Huang
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Yi Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Haojie Wu
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Wenhua Jian
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
| | - Zhangkai J Cheng
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou International Bio Island, XingDaoHuanBei Road, Guangzhou, 510005, Guangdong Province, China.
| | - Xiujing Han
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
| | - Baoqing Sun
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou International Bio Island, XingDaoHuanBei Road, Guangzhou, 510005, Guangdong Province, China.
| |
Collapse
|
126
|
Gao P, Zhou C, Yang H, Deng S, Jiang B, Wang Z, Di J, Su X. Comparison of circulating immune signatures across different consensus molecular subtypes (CMS) and between left- and right-sided lesions in stage II colorectal cancer using single-cell mass cytometry. J Transl Med 2025; 23:496. [PMID: 40312697 PMCID: PMC12044728 DOI: 10.1186/s12967-025-06481-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/11/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Patients with stage II colorectal cancer (CRC) show considerable variability in prognosis. Circulating immune cells play a vital role in systemic tumor surveillance. This study aimed to determine the clinical significance of the frequency and phenotype of circulating immune cell subsets in patients with stage II CRC. METHODS We applied a 37-marker-cell-lineage-agnostic panel to perform single-cell mass cytometry on peripheral blood mononuclear cells (PBMCs) from 73 patients with stage II CRC and 21 patients with stage III CRC. To categorize the stage II patients into consensus molecular subtypes (CMS), we performed RNA sequencing on tumor and adjacent normal tissues from 51 of the 73 patients. We then compared the immune cell phenotypes and frequencies based on tumor location and CMS classification in patients with stage II CRC. Wilcoxon test was employed to compare the mean frequencies of cell clusters between different tumor locations. Krustal-Wallis analysis with post-hoc Dunn test was used to assess differences across multiple CMS groups. RESULTS Stage II CRC patients with left- and right-sided tumors, as well as in different CMS groups, exhibit significantly different tumor characteristics. Single-cell mass cytometry revealed profound interpatient variability in immune cell subpopulation distribution and phenotypes in PBMCs in stage II CRC. We identified unique T:monocyte complexes in the peripheral blood of patients with stage II CRC, with significantly higher frequencies in these patients compared to those with stage III CRC. Left- and right-sided stage II CRCs differed in peripheral immunity. Patients with right-sided CRC had significantly higher frequencies of circulating CD8+CD27-CD28- immunosenescent T cell subsets compared to patients with left-sided CRC. Significant variations were observed in the subpopulations of the T:monocyte complex, T cells, and monocytes across different CMS groups Patients with CMS1 tumors (immune-active) exhibited significantly higher frequencies of CD4+ central memory and CD8+ terminal effector T cell: classical monocyte complexes, as well as CD8+ terminal effector T cells in the circulation. CONCLUSIONS The frequency and phenotype of circulating immune cells are influenced by tumor side and CMS subtypes in stage II CRC. These observations provide a basis for further investigation into the mechanisms linking tumors and systemic immunity.
Collapse
Affiliation(s)
- Pin Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Chuanyong Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Hong Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Shunyu Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Beihai Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Zaozao Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Jiabo Di
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| | - Xiangqian Su
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
127
|
Sena LA. Polyamine metabolism in prostate cancer. Curr Opin Oncol 2025; 37:223-232. [PMID: 40071465 PMCID: PMC11971019 DOI: 10.1097/cco.0000000000001134] [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: 04/05/2025]
Abstract
PURPOSE OF REVIEW Normal and malignant prostate engage in high rates of de novo polyamine synthesis. This review considers how polyamine metabolism regulates prostate cancer initiation and progression. RECENT FINDINGS The androgen receptor (AR) establishes a metabolic program to drive robust polyamine synthesis in the normal prostate. Upon malignant transformation, this AR-driven metabolic program persists and is optimized for oncogenesis by the proto-oncogene MYC and/or alterations to PI3K signaling. A deeper understanding of the function of polyamines in prostate cancer may be obtained by considering their function in the normal prostate. SUMMARY Recent findings support ongoing research into the role of polyamines in driving prostate cancer initiation and progression and suggest targeting polyamine metabolism remains a promising therapeutic strategy for prevention and treatment of prostate cancer.
Collapse
Affiliation(s)
- Laura A Sena
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, Maryland, USA
| |
Collapse
|
128
|
Sarrafan-Chaharsoughi Z, Yazdian Anari P, Malayeri AA, Naraev B, Del Rivero J. Update on Adrenocortical Carcinoma. Urol Clin North Am 2025; 52:275-286. [PMID: 40250894 DOI: 10.1016/j.ucl.2025.01.009] [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: 04/20/2025]
Abstract
Adrenocortical carcinoma (ACC) is an uncommon and highly aggressive form of cancer that originates from the adrenal glands. It displays a dual age distribution pattern, with a higher occurrence during early childhood around the average age of 3.2 years, and a second peak in the fourth and fifth decades of adulthood. The diagnosis of ACC requires a multifaceted approach, with accurate risk assessment being crucial for effective treatment planning. This review article offers a comprehensive overview of ACC, encompassing its epidemiology, clinical presentation, diagnosis, prognostic factors, and treatment methodologies.
Collapse
Affiliation(s)
| | - Pouria Yazdian Anari
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; Transitional Year Department, Garnet Health Medical Center, Middletown, New Work, NY 10940, USA
| | - Ashkan A Malayeri
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Boris Naraev
- Tampa General Hospital Cancer Institute, University of South Florida Morsani College of Medicine, Tampa, FL 33606, USA
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
129
|
Wang W, Li X, Wang H, Huang C, Zhu L, Wang H, Zhang W. CircDIAPH1 Promotes Liver Metastasis and Development of Colorectal Cancer by Initiation of CEACAM6 Expression. Mol Carcinog 2025; 64:897-910. [PMID: 39987565 DOI: 10.1002/mc.23896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/25/2025] [Accepted: 02/10/2025] [Indexed: 02/25/2025]
Abstract
Liver metastasis is a critical factor influencing the 5-year survival rate in colorectal cancer (CRC). However, the biological function of most circRNAs in liver metastasis of CRC is still unknown. In this study, we identified differentially expressed circRNAs associated with liver metastasis (LM-DE-circRNAs). A total of 247 LM-DE-circRNAs were identified, and crucial signaling pathways, including the regulation of actin cytoskeleton, were significantly enriched, featuring six LM-DE-circRNAs. Notably, circDIAPH1 (hsa_circ_0074323), with the highest AUC value, emerged as a potential biomarker for CRC liver metastasis (CRLM). Functional assays following circDIAPH1 knockdown demonstrated induced apoptosis, suppressed proliferation, reduced metastasis, and invasion in CRC cell lines in vitro. The circDIAPH1 knockdown attenuated tumor growth in a cell-derived xenograft model. Furthermore, circDIAPH1 knockdown lessened the liver metastasis. Transcriptome profiling revealed that CEACAM6 was the most downregulated gene while circDIAPH1 was knocked down, and possesses high expression value in CRC. Most importantly, we found that circDIAPH1 recruited transcription factor FOXA1 to bind in the promoter region of CEACAM6 and initiated CEACAM6 expression. Additionally, the study identified the transcription factor BRD4 as a regulator of circDIAPH1 expression in CRC. In conclusion, this study reveals that circDIAPH1 recruits FOXA1 to initiate CEACAM6 expression, promoting liver metastasis and development of CRC.
Collapse
Affiliation(s)
- Wei Wang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xu Li
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Hantao Wang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Cheng Huang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Laicheng Zhu
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Hao Wang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wei Zhang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
130
|
Rastrygina VA, Kazakov AS, Fadeev RS, Meshcheriakova EI, Deryusheva EI, Sokolov AS, Permyakova ME, Litus EA, Uversky VN, Permyakov EA, Permyakov SE. Soluble form of tumor necrosis factor-related apoptosis-inducing ligand interacts with S100P protein. Int J Biol Macromol 2025; 311:143667. [PMID: 40318722 DOI: 10.1016/j.ijbiomac.2025.143667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/22/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Tumor Necrosis Factor (TNF)-Related Apoptosis-Inducing Ligand (TRAIL) is a therapeutically relevant protein belonging to the TNF superfamily. Both membrane-bound and soluble (sTRAIL) forms of TRAIL affect innate and adaptive immune responses. We recently showed that soluble TNF binds specific members of the S100 family of multifunctional calcium-binding proteins, leading to suppression of its cytotoxic activity (Int. J. Mol. Sci. 2022, 23(24), 15,956). To test the ability of S100 proteins to affect sTRAIL functioning, we used surface plasmon resonance spectroscopy, intrinsic fluorescence, chemical crosslinking, molecular modeling, site-directed mutagenesis, cytotoxicity assay, and bioinformatics to study interaction of human sTRAIL with human non-fused S100 proteins. Of the 21 S100 proteins examined, only S100P protein showed specific interaction with sTRAIL characterized by equilibrium dissociation constant, Kd, reaching (0.16 ± 0.07) μM. sTRAIL monomer binds dimeric S100P strictly in the presence of Ca2+, while sTRAIL trimer interacts with S100P dimer regardless of Ca2+. Site-directed mutagenesis confirmed involvement of the 'hinge' and C-terminal regions of S100P in the sTRAIL recognition, consistent with the structural modeling results. Bioinformatic analysis indicates dysregulation of TRAIL and S100P in various neoplasms. S100P lowers cytotoxicity of sTRAIL against human fibrosarcoma HT-1080 cells. The suppression of proapoptotic sTRAIL signaling by S100P protein may contribute to oncogenic effects of the latter.
Collapse
Affiliation(s)
- Victoria A Rastrygina
- Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia
| | - Alexey S Kazakov
- Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia
| | - Roman S Fadeev
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Institutskaya str., 3, Pushchino, Moscow Region 142290, Russia
| | - Elena I Meshcheriakova
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Institutskaya str., 3, Pushchino, Moscow Region 142290, Russia
| | - Evgenia I Deryusheva
- Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia
| | - Andrey S Sokolov
- Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia
| | - Maria E Permyakova
- Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia
| | - Ekaterina A Litus
- Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Eugene A Permyakov
- Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia
| | - Sergei E Permyakov
- Institute for Biological Instrumentation, Institutskaya str., 7, Pushchino, Moscow Region 142290, Russia.
| |
Collapse
|
131
|
Sun Y, Wei H, Yu W, Gao H, Li J, Li X, Zhang H, Zhang H, Miao S, Zhao L, Yang R, Xu J, Lu Y, Wei F, Zhou H, Gao D, Jin Y, Zhang L. The actin-binding protein drebrin disrupts NF2-LATS kinases complex assembly to facilitate liver tumorigenesis. Hepatology 2025; 81:1433-1451. [PMID: 39325963 DOI: 10.1097/hep.0000000000001063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/17/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND AND AIMS The Hippo signaling has emerged as a crucial regulator of tissue homeostasis, regeneration, and tumorigenesis, representing a promising therapeutic target. Neurofibromin 2 (NF2), a component of Hippo signaling, is directly linked to human cancers but has been overlooked as a target for cancer therapy. APPROACH AND RESULTS Through a high-content RNA interference genome-wide screen, the actin-binding protein Drebrin (DBN1) has been identified as a novel modulator of YAP localization. Further investigations have revealed that DBN1 directly interacts with NF2, disrupting the activation of large tumor suppressor kinases (LATS1/2) by competing with LATS kinases for NF2 binding. Consequently, DBN1 knockout considerably promotes YAP nuclear exclusion and repression of target gene expression, thereby preventing cell proliferation and liver tumorigenesis. We identified three lysine residues (K238, K248, and K252) essential for DBN1-NF2 interaction and developed a mutant DBN1 (DBN1-3K mut ) that is defective in NF2 binding and incompetent to trigger NF2-dependent YAP activation and tumorigenesis both in vitro and in vivo. Furthermore, BTP2, a DBN1 inhibitor, successfully restored NF2-LATS kinase binding and elicited potent antitumor activity. The combination of sorafenib and BTP2 exerted synergistic inhibitory effects against HCC. CONCLUSIONS Our study identifies a novel DBN1-NF2-LATS axis, and pharmacological inhibition of DBN1 represents a promising alternative intervention targeting the Hippo pathway in cancer treatment.
Collapse
Affiliation(s)
- Yang Sun
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Henan Wei
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Wentao Yu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Haoran Gao
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jinhui Li
- HuidaGene Therapeutics Co., Ltd., Shanghai, China
| | - Xiaoyu Li
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Haijiao Zhang
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Haoen Zhang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Sen Miao
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Lihua Zhao
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Ruizeng Yang
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Jinjin Xu
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Yi Lu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Fang Wei
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Hu Zhou
- State Key Laboratory of Drug Research, Department of Analytical Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Daming Gao
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yunyun Jin
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Lei Zhang
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
132
|
Lu S, Zhou Q, Zhao R, Xie L, Cao WM, Feng YX. Unraveling UPR-mediated intercellular crosstalk: Implications for immunotherapy resistance mechanisms. Cancer Lett 2025; 617:217613. [PMID: 40054654 DOI: 10.1016/j.canlet.2025.217613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/10/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Endoplasmic reticulum (ER) is the critical organelle that regulates essential cellular processes, including protein synthesis, folding, and post-translational modification, as well as lipid metabolism and calcium homeostasis. Disruption in ER homeostasis leads to a condition known as ER stress, characterized by the accumulation of misfolded or unfolded proteins. This triggers the unfolded protein response (UPR), an adaptive pathway mediated by three ER-resident sensors: inositol-requiring enzyme 1α (IRE1α), protein kinase R-like ER kinase (PERK), and activating transcription factor 6 (ATF6). Increasing evidence highlights sustained UPR activation in malignant and immune cells within the tumor microenvironment (TME), which promotes tumor progression and metastasis while simultaneously impairing antitumor immunity. This review explores how UPR-driven intercellular signaling influences immunotherapy resistance, focusing on the alterations occurring in tumor cells as well as in the surrounding immune environment. By providing insights into these mechanisms, we aim to highlight the therapeutic potential of targeting the UPR pathways in modulating cancer immunity.
Collapse
Affiliation(s)
- Si Lu
- Department of Head and Neck Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China; Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, China
| | - Qimin Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rongjie Zhao
- Department of Gynecological Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| | - Lei Xie
- Department of Head and Neck Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wen-Ming Cao
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China.
| | - Yu-Xiong Feng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China; Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, China.
| |
Collapse
|
133
|
Tuo Z, Gao M, Jiang C, Zhang D, Chen X, Jiang Z, Wang J. Construction of M2 macrophage-related gene signature for predicting prognosis and revealing different immunotherapy response in bladder cancer patients. Clin Transl Oncol 2025; 27:2191-2206. [PMID: 39347941 DOI: 10.1007/s12094-024-03698-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/22/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Bladder cancer development is closely associated with the dynamic interaction and communication between M2 macrophages and tumor cells. However, specific biomarkers for targeting M2 macrophages in immunotherapy remain limited and require further investigation. METHODS In this study, we identified key co-expressed genes in M2 macrophages and developed gene signatures to predict prognosis and immunotherapy response in patients. Public database provided the bioinformatics data used in the analysis. We created and verified an M2 macrophage-related gene signature in these datasets using Lasso-Cox analysis. RESULTS The predictive value and immunological functions of our risk model were examined in bladder cancer patients, and 158 genes were found to be significantly positively correlated with M2 macrophages. Moreover, we identified two molecular subgroups of bladder cancer with markedly different immunological profiles and clinical prognoses. The five key risk genes identified in this model were validated, including CALU, ECM1, LRP1, CYTL1, and CCDC102B, demonstrating the model can accurately predict prognosis and identify unique responses to immunotherapy in patients with bladder cancer. CONCLUSIONS In summary, we constructed and validated a five-gene signature related to M2 macrophages, which shows strong potential for forecasting bladder cancer prognosis and immunotherapy response.
Collapse
Affiliation(s)
- Zhouting Tuo
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Mingzhu Gao
- Department of Oncology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Chao Jiang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Duobing Zhang
- Department of Urology, Suzhou Hospital of Anhui Medical University, Suzhou, 234000, China
- Department of Urology, Suzhou Municipal Hospital of Anhui Province, Suzhou, 234000, China
| | - Xin Chen
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhiwei Jiang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Jinyou Wang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
134
|
Zhao Z, Shi Y, Chen S, Xu Y, Fu F, Li C, Zhang X, Li M, Li X. Development of a web-based tool for estimating individualized survival curves in glioblastoma using clinical, mRNA, and tumor microenvironment features with fusion techniques. Clin Transl Oncol 2025; 27:2113-2126. [PMID: 39476302 DOI: 10.1007/s12094-024-03739-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/16/2024] [Indexed: 04/27/2025]
Abstract
OBJECTIVE Glioblastoma (GBM), one of the most common brain tumors, is known for its low survival rates and poor treatment responses. This study aims to create a robust predictive model that integrates multiple feature types, including clinical data, RNA expression, and tumor microenvironment data, using fusion techniques to enhance model performance. METHODS We obtained data from the SEER database to assess the impact of nine demographic and clinical features on the survival of 58,495 GBM patients and built predictive machine learning models. Additionally, mRNA expression data from 600 GBM patients from TCGA, CGGA, and GEO were analyzed. We used Cox regression and LASSO to create a gene signature, which was compared against 13 published signatures for accuracy. Twenty-one machine learning models were applied to predict survival at multiple time points. Finally, we integrated multiple feature types using fusion techniques and developed a Shiny app to provide survival predictions for GBM patients. RESULTS Using the SEER database, we constructed machine learning models based on nine clinical variables: age, gender, marital status, race, tumor site, laterality, surgery, chemotherapy, and radiation therapy. The best-performing model achieved AUC values of 0.775, 0.728, 0.692, and 0.683 for predicting survival at 6, 12, 18, and 24 months in the testing cohort. In the merged TCGA, CGGA, and GEO cohorts, we identified 11 genes to develop predictive models. These 11 genes outperformed 13 other published gene signatures in predicting the prognosis of GBM. When incorporating mRNA features, tumor microenvironment features, and clinical variables into the fusion models, the AUC values for predicting survival at 6, 12, 18, and 24 months were 0.641, 0.624, 0.655, and 0.637, respectively. A user-friendly tool for predicting the survival curve of individual GBM patients is available at https://zzubioinfo.shinyapps.io/mlGBM/ . CONCLUSIONS Our study provides a web-based tool that includes two modules: one for predicting survival curves using only clinical variables, and another that integrates multiple feature types for more comprehensive predictions.
Collapse
Affiliation(s)
- Zunlan Zhao
- Department of General Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yujie Shi
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shouhang Chen
- Department of Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan, China
| | - Yan Xu
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Fangfang Fu
- Department of Medical Imaging, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chong Li
- Department of Pharmacy, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ming Li
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China.
| | - Xiqing Li
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
135
|
Song G, Zeng C, Li J, Liu J, Zhao J, Liu B, Fan J, Xie H. Exosome-based nanomedicines for digestive system tumors therapy. Nanomedicine (Lond) 2025; 20:1167-1180. [PMID: 40248953 DOI: 10.1080/17435889.2025.2493037] [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: 01/18/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025] Open
Abstract
Digestive system tumors constitute a major subset of malignancies, consistently ranking among the leading causes of mortality globally. Despite limitations inherent in current therapeutic modalities, recent advancements in targeted therapy and drug delivery systems have led to significant improvements in the efficacy of pharmacotherapy for digestive system tumors. In this context, exosomes - naturally occurring nanoscale vesicles - have emerged as promising drug delivery candidates due to their intrinsic molecular transport capabilities, superior biocompatibility, and targeted recognition of tumor cells. The integration of exosomes into cancer therapeutics represents a novel and potentially transformative approach for treating digestive system tumors, which may drive further progress in this field. This review comprehensively examines the sources, loading mechanisms, and therapeutic efficacy of exosomes in the context of digestive system tumor treatment. Furthermore, it discusses the opportunities and challenges associated with exosomes, offering insights into their future role within the therapeutic armamentarium against digestive tumors.
Collapse
Affiliation(s)
- Ge Song
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Chenlu Zeng
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Junru Li
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Jiajia Liu
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Juanxia Zhao
- Department of Pathology, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, Hunan, China
| | - Jialong Fan
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
| | - Hailong Xie
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| |
Collapse
|
136
|
Ebrahim NAA, Othman MO, Salama RA, Abdelfatah D, Tahoun NS. Diagnostic insights into solid pseudopapillary neoplasms of the pancreas: a decade of experience with pediatric representation. Diagn Pathol 2025; 20:57. [PMID: 40307756 PMCID: PMC12042314 DOI: 10.1186/s13000-025-01648-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Solid pseudopapillary neoplasms (SPNs) of the pancreas are rare, low-grade malignancies that predominantly affect young females. Their diagnosis is often facilitated by a characteristic histomorphological pattern and immunohistochemical profile. However, diagnostic challenges persist, especially in pediatric and atypical presentations. Recent attention has focused on the diagnostic value of CD99 and LEF1 in distinguishing SPNs from other pancreatic neoplasms. OBJECTIVE To evaluate the diagnostic accuracy and utility of CD99 and LEF1 as immunohistochemical markers for SPNs. METHODS A retrospective analysis of 60 SPN cases diagnosed between 2015 and 2024 was performed. Histopathological features were systematically reviewed, and immunohistochemical staining for CD99, LEF1, β-catenin, Cyclin D1, PR, Ki-67 was evaluated. Immunohistochemical marker interpretation was standardized using internally validated thresholds informed by existing literature: CD99 was considered positive with ≥ 10% cytoplasmic staining exhibiting paranuclear accentuation; β-catenin positivity was defined by ≥ 5% nuclear localization; Cyclin D1 by ≥ 10% moderate-to-strong nuclear staining; and progesterone receptor (PR) expression by ≥ 1% nuclear positivity, consistent with hormone receptor evaluation guidelines. Marker expression was statistically analyzed for their associations. RESULTS SPNs exhibited a strong female predilection (F:M ratio ≈ 7:1), with a mean age of 32.5 years. Pediatric cases (n = 4) displayed higher mean expression of CD99 (73.8%) and LEF1 (86.5%) compared to adults. CD99 showed cytoplasmic positivity with paranuclear accentuation in 96.7% of cases, while LEF1 demonstrated nuclear staining in 91.7%. β-catenin nuclear localization was observed in 95% of tumors, reflecting Wnt/β-catenin pathway activation. Cyclin D1 and PR were expressed in 90% and 88.3% of cases, respectively. Co-expression of β-catenin, CD99, LEF1, Cyclin D1, and PR was observed in 73.3% of tumors. CD99 and LEF1 inversely correlated with tumor size and proliferative activity (Ki-67), whereas Cyclin D1 and Ki-67 positively correlated with tumor size and lymphovascular invasion (LVI). Pediatric tumors generally exhibited favorable profiles, with limited evidence of LVI. CONCLUSION SPNs present with distinctive immunohistochemical signatures that are critical for accurate diagnosis, particularly in morphologically ambiguous or pediatric cases. CD99 and LEF1 are highly sensitive markers that, in combination with β-catenin and Cyclin D1, enhance diagnostic precision. These findings emphasize the central role of Wnt/β-catenin signaling in SPN pathogenesis and underscore the importance of integrating clinicopathological and molecular data for comprehensive tumor assessment.
Collapse
Affiliation(s)
- Noura A A Ebrahim
- Oncologic Pathology Department, National Cancer Institute (NCI) - Cairo University, Cairo, Egypt.
| | - Moamen O Othman
- Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rasha A Salama
- Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE
| | - Dalia Abdelfatah
- Cancer Epidemiology and Biostatistics Department, National Cancer Institute (NCI) - Cairo University, Cairo, Egypt
| | - Neveen S Tahoun
- Oncologic Pathology Department, National Cancer Institute (NCI) - Cairo University, Cairo, Egypt
| |
Collapse
|
137
|
Zhang Y, He H, Fu X, Liu G, Wang H, Zhong W, Xu X, Chen B, Mei L. Glioblastoma-associated macrophages in glioblastoma: from their function and mechanism to therapeutic advances. Cancer Gene Ther 2025:10.1038/s41417-025-00905-9. [PMID: 40307579 DOI: 10.1038/s41417-025-00905-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor in adults and has high mortality rates worldwide. GBM progression, treatment, and prognosis are influenced by the tumor microenvironment (TME), which includes immune, stromal, and tumor cells. Among them, glioblastoma-associated macrophages (GAMs) act as key regulators of GBM pathobiology. GAMs exhibit remarkable plasticity, as they can exhibit both protumor and antitumor effects. However, their function is determined by polarization and the TME. In this review, we provide a comprehensive overview of the current understanding of the biology of GAMs in GBM, including their origins, phenotypic diversity, and functional roles. We discuss the intricate crosstalk between GAMs and tumor cells, as well as other immune and stromal components, and highlight the mechanisms underlying GAM-mediated tumor progression, invasion, angiogenesis, and immune system evasion. Furthermore, we explore the therapeutic implications of targeting GAMs in GBM and discuss emerging strategies aimed at reprogramming GAMs toward an antitumorigenic phenotype or selectively depleting protumorigenic subsets. The final aim is to develop innovative therapeutic approaches that disrupt GBMs. By leveraging our increased understanding of GAM biology, we lay the foundation for transformative advances in GBM treatment to improve patient prognosis.
Collapse
Affiliation(s)
- Yuqin Zhang
- Department of General Practice, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hanxing He
- Department of Orthopedics and Traumatology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - Xin Fu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Ganzhi Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huiying Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen Zhong
- Department of General Practice, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xia Xu
- Department of General Practice, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Bo Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China.
| | - Lin Mei
- Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin, China.
| |
Collapse
|
138
|
Martínez-Ríos J, López-Pacheco CP, García-Zepeda EA, Soldevila G. CCR9 shapes the immune microenvironment of colorectal cancer modulating the balance between intratumoral CD8+ T cell and FoxP3+ Helios+ Treg subpopulations. PLoS One 2025; 20:e0321930. [PMID: 40305493 PMCID: PMC12043142 DOI: 10.1371/journal.pone.0321930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/12/2025] [Indexed: 05/02/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world and the second cause of death related to cancer. Regulatory T cell (Treg) infiltration is enriched in several tumor types including CRC and correlates with suppression of the anti-tumor immune response. In the large intestine, thymic Tregs (tTregs Helios+) and peripheral Tregs (pTregs Helios-) coexist and maintain intestinal homeostasis under steady state conditions. The recruitment of Treg cells to the intestine is orchestrated by the CCR9/CCL25 axis, which is potentiated under inflammatory conditions. Interestingly, the balance between cytotoxic CD8+ T cells and Tregs within the tumor microenvironment is critical for antitumor immunity and cancer progression. An elevated CD8+/Treg ratio has been associated with improved clinical outcomes in various cancers, including CRC. Therefore, here we investigate the potential role of chemokine receptor CCR9 on CD8+/Treg ratio and the effect of the recruitment of Treg subpopulations (Helios+ and Helios-) into the tumor microenvironment using the AOM/DSS induced colitis-associated colorectal cancer murine model. Our findings reveal that CCR9 deficiency leads to distinct alterations in the CRC microenvironment, characterized by decreased intratumoral Tregs Helios+. Also, the lack of the receptor leads to an improvement of the antitumor immune response, increasing the CD8+/Treg ratio within the tumor immune infiltrate. These results underscore the importance of CCR9 in shaping the immune microenvironment during CRC development.
Collapse
Affiliation(s)
- Jacobo Martínez-Ríos
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Cynthia Paola López-Pacheco
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Investigadora por México, Secretaría de Ciencia Tecnología y Humanidades (SECIHTI), Mexico City, Mexico
| | - Eduardo Alberto García-Zepeda
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gloria Soldevila
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio Nacional de Citometría de Flujo, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
139
|
Yin H, Shi J, Li S, You Q, Zhu H, Koo C, Liu B, Hou L, Wu C. Emerging roles of exosomal circRNAs in non-small cell lung cancer. J Transl Med 2025; 23:490. [PMID: 40307927 PMCID: PMC12042431 DOI: 10.1186/s12967-025-06463-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: 02/13/2025] [Accepted: 04/06/2025] [Indexed: 05/02/2025] Open
Abstract
Despite the prevalence of non-small cell lung cancer (NSCLC) is high, the limited early detection and management of these tumors are restricted since there is an absence of reliable and precise diagnostic biomarkers and therapeutic targets. Exosomes transport functional molecules for facilitating intercellular communication, especially in the tumor microenvironment, indicating their potential as cancer biomarkers and therapeutic targets. Circular RNA (circRNA), a type of non-coding RNA possessing a covalently closed loop structure, substantial abundance, and tissue-specific expression patterns, is stably enriched in exosomes. In recent years, significant breakthroughs have been made in research on exosomal circRNA in NSCLC. This review briefly introduces the biogenesis, characterizations, and functions of circRNAs and exosomes, and systematically describes the biological functions and mechanisms of exosomal circRNAs in NSCLC. In addition, this study summarizes their role in the progression of NSCLC and discusses their clinical significance as biomarkers and therapeutic targets for NSCLC.
Collapse
Affiliation(s)
- Hongyuan Yin
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jiayi Shi
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shaoling Li
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Qianhui You
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huici Zhu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chinying Koo
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Baonian Liu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Likun Hou
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| |
Collapse
|
140
|
McDermott A, Juárez M, Wade MG, Patten SA, Plante I. Exposure to brominated flame retardants during pregnancy and lactation increases the prevalence of breast lesions and cancer-associated pathways in sprague-dawley rats. Reprod Toxicol 2025; 135:108928. [PMID: 40316034 DOI: 10.1016/j.reprotox.2025.108928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/01/2025] [Accepted: 04/21/2025] [Indexed: 05/04/2025]
Abstract
The mammary gland undergoes significant changes during pregnancy, lactation, and involution, making it highly susceptible to endocrine-disrupting chemicals such as brominated flame retardants (BFRs). Despite being restricted in many countries, some BFRs persist in the environment and accumulate in human tissues, including the mammary gland and human milk. This study investigates the effects of BFRs exposure during pregnancy and lactation on mammary gland development and breast cancer risk in a rat model. Dams were exposed to a mixture of polybrominated diphenyl ethers (PBDEs) and hexabromocyclododecane (HBCDD), formulated based on relative congener levels found in house dust. Post-weaning, dams were treated with 7,12-dimethylbenz[a]anthracene (DMBA) to initiate tumor formation. The results revealed that both low and high doses of BFRs induced lesions in mammary epithelium, with an increase in total lesion number in low dose. Molecular analysis revealed disruptions in the Wnt/β-catenin signaling pathway, leading to an increase in oncogene expression, including c-Myc and c-Jun. RNA sequencing also indicated dysregulation in calcium signaling and glucose metabolism pathways. Our findings suggest that BFR exposure during the critical window of mammary gland involution compromises the cancer-protective effects of pregnancy and lactation. These effects are particularly significant at low exposure levels, demonstrating a non-monotonic dose-response. The study underscores the potential long-term health risks associated with environmental BFR exposure and highlights the need for further research on its implications on the risks of developing breast cancer later in life.
Collapse
Affiliation(s)
- Alec McDermott
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Melany Juárez
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Michael G Wade
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, Canada
| | | | - Isabelle Plante
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada.
| |
Collapse
|
141
|
Zhou Q, Song B, He Y, Zhang Z, Chen S, Chen W, Li X, Jiang J. Identification of a disulfidptosis-related genes signature for diagnostic and immune infiltration characteristics in cervical cancer. PLoS One 2025; 20:e0322387. [PMID: 40305445 PMCID: PMC12043181 DOI: 10.1371/journal.pone.0322387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/17/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Cervical cancer (CC) ranks as the fourth most common malignancy affecting women globally, with research highlighting a rising incidence among younger age groups. Disulfidptosis, a newly identified form of regulated cell death, has been implicated in the pathogenesis of numerous diseases. This study employs bioinformatics analyses to explore the expression profiles and functional roles of disulfidptosis-related genes (DRGs) in the context of cervical cancer. METHODS Differential analysis of the gene expression matrix in CC was performed to identify differentially expressed genes. The overlap between these genes and disulfidptosis-related genes was then determined. Key hub genes were identified using multiple machine learning approaches, including LASSO regression, support vector machines (SVM), and random forest (RF). These hub genes were subsequently used to construct a predictive model, which was validated using external datasets to ensure robustness and reliability. RESULTS In this study, 11 overlapping genes were identified, among which four hub genes-BRK1, NDUFA11, RAC1, and NDUFS1-were extracted using machine learning techniques. The diagnostic performance of these hub genes was validated with external datasets, and a predictive model was constructed based on their expression. The model demonstrated an exceptionally high area under the curve (AUC) of 0.997. Moreover, AUC values exceeding 0.85 for two independent validation datasets further confirmed the model's accuracy and stability. Notably, NDUFA11 and BRK1 showed significant associations with patient survival, highlighting their prognostic importance in cervical squamous cell carcinoma. Using CMAP and DGIdb databases, Metformin and Coenzyme-I were identified as potential targeted therapies for NDUFS1 and NDUFA11, respectively, offering new therapeutic avenues for patients. CONCLUSION This study uncovered a strong association between disulfidptosis and CC and developed a predictive model to assess the risk in CC patients. These findings offer novel insights into identifying biomarkers and potential therapeutic targets for CC, paving the way for improved diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Qun Zhou
- Department of gynecology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Bangli Song
- Department of Internal Medicine, Zhejiang University of Technology Hospital, Hangzhou, Zhejiang, China
| | - Yibo He
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, ZheJiang, China
| | - Zhezhong Zhang
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, ZheJiang, China
| | - Shiliang Chen
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, ZheJiang, China
| | - Wenjun Chen
- School of Nursing, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xianbin Li
- School of Computer and Big Data Science, Jiujiang University, Jiujiang, China
| | - Jun Jiang
- Department of gynecology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
142
|
Hadri SH, Riaz A, Abid J, Shaheen R, Nadeem S, Ghumman Z, Naeem H. Emerging nanostructure-based strategies for breast cancer therapy: innovations, challenges, and future directions. Med Oncol 2025; 42:188. [PMID: 40307624 DOI: 10.1007/s12032-025-02743-z] [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: 01/25/2025] [Accepted: 04/23/2025] [Indexed: 05/02/2025]
Abstract
Breast cancer, one of the leading causes of cancer-associated deaths, is responsible for the majority of cases of cancer in women globally. Traditional therapies used for the treatment of cancer have some challenges such as low cellular absorption, multidrug resistance, and limited bioavailability. Current innovations in nanotechnology, such as nanoliposomes, silver nanoparticles, gold nanoparticles, and carbon nanotubes, provide a promising approach to deal with these limitations. Nanostructures encapsulating anticancer agents such as doxorubicin, curcumin, paclitaxel, erlotinib, and docetaxel enhance the therapeutic efficacy of these agents and promote targeted drug delivery. Curcumin-loaded amorphous calcium carbonate nanoparticles encapsulating lipids and L-arginine exhibit higher cytotoxicity than free curcumin. Gold nanoparticles can also enhance treatment efficacy by specifically destroying tumor cells when used in photothermal therapy. This review focus on the abilities of nanoparticles to induce oxidative stress, prevent proliferation, and trigger apoptosis in cancer cells. Further research should focus on optimizing these nanoparticles to enhance the targeted drug delivery and address multi-drug resistance. Our review underscores recent developments in nanostructures, their therapeutic potential, and the challenges that need to be addressed for more effective breast cancer treatment.
Collapse
Affiliation(s)
- Saqib Hussain Hadri
- Department of Biochemistry and Biotechnology, University of Gujrat, Hafiz Hayat Campus, Gujrat, Pakistan
| | - Atiya Riaz
- Department of Biochemistry and Biotechnology, University of Gujrat, Hafiz Hayat Campus, Gujrat, Pakistan.
| | - Jaisha Abid
- Department of Biochemistry and Biotechnology, University of Gujrat, Hafiz Hayat Campus, Gujrat, Pakistan
| | - Rameeza Shaheen
- Department of Biochemistry and Biotechnology, University of Gujrat, Hafiz Hayat Campus, Gujrat, Pakistan
| | - Samreen Nadeem
- Department of Biochemistry and Biotechnology, University of Gujrat, Hafiz Hayat Campus, Gujrat, Pakistan
| | - Zainab Ghumman
- Department of Biochemistry and Biotechnology, University of Gujrat, Hafiz Hayat Campus, Gujrat, Pakistan
| | - Hammad Naeem
- Department of Biochemistry and Biotechnology, University of Gujrat, Hafiz Hayat Campus, Gujrat, Pakistan
| |
Collapse
|
143
|
Yi K, Huang Y, Jiang Y, Zhou L. Causal relationship between gut microbiota and laryngeal cancer: a mendelian randomization analysis. Braz J Otorhinolaryngol 2025; 91:101634. [PMID: 40305979 DOI: 10.1016/j.bjorl.2025.101634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
OBJECTIVE Laryngeal cancer incidence is rising globally; the role of gut microbiota remains underexplored. This study aimed to establish a causal link between gut microbiota and laryngeal cancer to inform preventive and therapeutic strategies. METHODS Gut microbiota data from GWAS conducted by the MiBioGen consortium served as the exposure variable, with laryngeal cancer as the outcome variable. the exposure variable and the outcome variable were analyzed using Mendelian Randomization. The primary method was Inverse Variance Weighted analysis, with heterogeneity and pleiotropy assessed through Cochran's Q test, MR-Egger regression, and MR-PRESSO. RESULTS In the study, we identified five bacterial taxa with potential causal relationships with laryngeal cancer risk: Higher levels of Clostridiaceae1 (OR = 0.9993, 95% CI 0.9986-0.9999, p = 0.0463) and Turicibacter (OR = 0.9995, 95% CI 0.9989-0.9999, p = 0.0384) were linked to reduced cancer risk, while Mollicutes RF9 (OR = 1.0010, 95% CI 1.0003-1.0016, p = 0.0027), Euryarchaeota (OR = 1.0004, 95% CI 1.0001-1.0007, p = 0.0234), and Cyanobacteria (OR = 1.0005, 95% CI 1.0000-1.0009, p = 0.0464) were associated with increased risk. CONCLUSION Our findings suggest a causal relationship between gut microbiota composition and laryngeal cancer risk. Clostridiaceae1 and Turicibacter may play a protective role, while Mollicutes RF9, Euryarchaeota, and Cyanobacteria could contribute to increased cancer susceptibility. These insights highlight potential microbiome-based strategies for early detection, prevention, and therapeutic intervention in laryngeal cancer. LEVEL OF EVIDENCE Level 5.
Collapse
Affiliation(s)
- Kaiyan Yi
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Yu Huang
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Yun Jiang
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Lingling Zhou
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
144
|
Han D, Wu C, Jin H. Anti-tumor immune modulation and favorable survival outcomes in uterine corpus endometrial carcinoma: insights from PIK3CA/ARID1A co-mutation analysis. Discov Oncol 2025; 16:641. [PMID: 40301230 PMCID: PMC12040806 DOI: 10.1007/s12672-025-02422-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/17/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Uterine corpus endometrial carcinoma (UCEC) is the most prevalent cancer of the female reproductive system, posing significant risks to women's reproductive health and imposing considerable economic burdens on families and society due to high treatment costs. METHODS The study population comprised 529 UCEC patients who were selected and retrieved from the cBioPortal public database for a comprehensive integrated analysis. This study aims to explore the prognostic significance of co-mutation in PIK3CA/ARID1A genes in UCEC, utilizing various bioinformatics approaches, including differential expression genes (DEGs) analysis, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, Gene Set Enrichment Analysis (GSEA), immune infiltration analysis, and the establishment of nomogram model. RESULTS PIK3CA/ARID1A co-mutation group had a better prognosis than the other three groups. The co-mutation of PIK3CA/ARID1A was associated with a significantly improved overall survival (OS) in patients with UCEC and immunotherapy markers. This result was further corroborated in the MSK cohort, reinforcing the robustness of our observations. Our findings revealed that 222 genes were upregulated and 1,464 genes downregulated in the co-mutation group compared to the non-co-mutation (NCM) group, providing a molecular basis for understanding the biological roles of these gene mutations in UCEC. Additionally, pathway analysis identified significant enrichment in immune-related pathways, emphasizing the potential for co-mutation to influence tumor progression via immune modulation. Notably, patients with co-mutations exhibited improved overall survival (P < 0.05), suggesting their role as vital prognostic markers. The developed Cox proportional hazards model demonstrated high predictive accuracy (C-index = 0.835), supporting personalized management for UCEC patients. CONCLUSION In conclusion, this study underscores the importance of PIK3CA and ARID1A co-mutations in UCEC, advocating for their further exploration in clinical applications and therapeutic strategies.
Collapse
Affiliation(s)
- Dongmei Han
- Center for Precision Cancer Medicine and Translation Research, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300181, China
- Center for Precision Cancer Medicine and Translation Research, Tianjin Medical University Cancer Institute & Hospital, No. 99, East 5 th Road, Tianjin Airport Economic Zone, Tianjin, 300060, China
| | - Caihong Wu
- Center for Precision Cancer Medicine and Translation Research, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300181, China
- Center for Precision Cancer Medicine and Translation Research, Tianjin Medical University Cancer Institute & Hospital, No. 99, East 5 th Road, Tianjin Airport Economic Zone, Tianjin, 300060, China
| | - Hao Jin
- Center for Precision Cancer Medicine and Translation Research, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300181, China.
- Center for Precision Cancer Medicine and Translation Research, Tianjin Medical University Cancer Institute & Hospital, No. 99, East 5 th Road, Tianjin Airport Economic Zone, Tianjin, 300060, China.
- Clinical Research Management Department, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300181, China.
| |
Collapse
|
145
|
Norouzkhani N, Mobaraki H, Varmazyar S, Zaboli H, Mohamadi Z, Nikeghbali G, Bagheri K, Marivany N, Najafi M, Nozad Varjovi M, Abouzeid M, Zeidi Baghrabad H, Eini P, Azhdarimoghaddam A, Khosravi F, Asadi Anar M. Artificial intelligence networks for assessing the prognosis of gastrointestinal cancer to immunotherapy based on genetic mutation features: a systematic review and meta-analysis. BMC Gastroenterol 2025; 25:310. [PMID: 40301768 PMCID: PMC12039286 DOI: 10.1186/s12876-025-03884-1] [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: 01/18/2025] [Accepted: 04/10/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND AND AIM Artificial intelligence (AI) networks offer significant potential for predicting immunotherapy outcomes in gastrointestinal cancers by analyzing genetic mutation profiles. Their application in prognosis remains underexplored. This systematic review and meta-analysis aim to evaluate the effectiveness of AI-based models, which refers to systems utilizing artificial intelligence to analyze data and make predictions, in predicting immunotherapy responses in gastrointestinal cancers using genetic mutation features. METHODS This study, adhering to PRISMA guidelines, aimed to evaluate AI networks for predicting gastrointestinal cancer prognosis in response to immunotherapy using genetic mutation features. A search in PubMed, WOS, and Scopus identified relevant studies. Data extraction and quality assessment were conducted, and statistical analysis included pooled estimates for sensitivity, specificity, accuracy, and AUC. Regression models and imputation methods addressed missing values, ensuring accurate and robust results. STATA version 18 was used to analyze the data. RESULT A total of 45 studies, all published in 2024, involving 14,047 participants in training sets and 10,885 participants in test sets, were included. The pooled results of AI model performance for gastrointestinal cancers based on genetic mutation features were: AUC = 0.86 (95% CI: 0.86-0.87), Sensitivity = 83% (95% CI: 83%-84%), Specificity = 72% (95% CI: 72%-73%), and Accuracy = 82% (95% CI: 82%-83%). Heterogeneity was low to moderate, and no publication bias was detected. Subgroup analysis showed higher AUC for gastric cancer models (AUC: 0.87) and lower for pancreatic cancer models (AUC: 0.52). CONCLUSION AI networks demonstrate promising potential in predicting immunotherapy outcomes for gastrointestinal cancers based on genetic mutation features. This systematic review highlights their effectiveness in stratifying patients and optimizing treatment decisions. However, further large-scale studies are needed to validate AI models and integrate them into clinical practice for improved precision in cancer immunotherapy.
Collapse
Affiliation(s)
- Narges Norouzkhani
- Department of Medical Informatics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hesam Mobaraki
- Faculty of Medicine, İstanbul Yeniyuzyil University, Istanbul, Turkey
| | | | - Hadis Zaboli
- Student Research Committee, Babol University Medical Science, Babol, Iran
| | - Zhina Mohamadi
- Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Golnaz Nikeghbali
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamyar Bagheri
- Student Research Committee, Abadan University of Medical Sciences, Abadan, Iran
| | - Newsha Marivany
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mirmehdi Najafi
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahdiyeh Nozad Varjovi
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohamed Abouzeid
- Mersey & West Lancashire Teaching Hospitals NHS Trust, Southport Hospital, Lancashire, UK.
| | - Hanieh Zeidi Baghrabad
- Tabriz University of Medical Sciences, Gastroenterology and Hepatology Research Center, Tabriz, Iran
| | - Pooya Eini
- Toxicological Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aida Azhdarimoghaddam
- Student Research Committee, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Farbod Khosravi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
146
|
Delmas D, Mialhe A, Cotte AK, Connat JL, Bouyer F, Hermetet F, Aires V. Lipid metabolism in cancer: Exploring phospholipids as potential biomarkers. Biomed Pharmacother 2025; 187:118095. [PMID: 40311223 DOI: 10.1016/j.biopha.2025.118095] [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: 02/12/2025] [Revised: 04/03/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Aberrant lipid metabolism is increasingly recognized as a hallmark of cancer, contributing to tumor growth, metastatic dissemination, and resistance to therapy. Cancer cells reprogram key metabolic pathways-including de novo lipogenesis, lipid uptake, and phospholipid remodeling-to sustain malignant progression and adapt to microenvironmental demands. This review summarizes current insights into the role of lipid metabolic reprogramming in oncogenesis and highlights recent advances in lipidomics that have revealed cancer type- and stage-specific lipid signatures with diagnostic and prognostic relevance. We emphasize the dual potential of lipid metabolic pathways-particularly those involving phospholipids-as sources of clinically relevant biomarkers and therapeutic targets. Enzymes and transporters involved in these pathways have emerged as promising candidates for both diagnostic applications and pharmacological intervention. We also examine persistent challenges hindering the clinical translation of lipid-based approaches, including analytical variability, insufficient biological validation, and the lack of standardized integration into clinical workflows. Furthermore, the review explores strategies to overcome these barriers, highlighting the importance of incorporating lipidomics into multi-omics frameworks, supported by advanced computational tools and AI-driven analytics, to decipher the complexity of tumor-associated metabolic networks. We discuss how such integrative approaches can facilitate the identification of actionable metabolic targets, improve the specificity and robustness of lipid-based biomarkers, and enhance patient stratification in the context of precision oncology.
Collapse
Affiliation(s)
- Dominique Delmas
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France; Centre de Lutte Contre le Cancer Georges François Leclerc Center, Dijon F-21000, France; Inserm UMS58 - Biologie Santé Dijon (BioSanD), Dijon F-21000, France.
| | - Aurélie Mialhe
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Alexia K Cotte
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Jean-Louis Connat
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Florence Bouyer
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - François Hermetet
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Virginie Aires
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| |
Collapse
|
147
|
Vu NN, Ng KW, Jaitpal S, Negahdary M, Nguyen T, Kodam RS, Mabbott S. High Precision Automated Synthesis of Surface-Enhanced (Resonance) Raman Nanotags. ACS Sens 2025. [PMID: 40299759 DOI: 10.1021/acssensors.5c00068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Batch-to-batch inconsistencies and time-intensive protocols remain significant challenges in conventional nanomaterial synthesis. Here, we present an automated system that precisely fabricates silica-coated gold nanostars (AuNS@SiO2) incorporating Raman reporters (4-MBA and IR-780) defined as nanotags, thereby enabling control over their morphological, optical, and spectroscopic properties. The resulting nanotags were comprehensively characterized through UV-vis spectrophotometry, transmission electron microscopy (TEM), surface enhanced (resonance) Raman spectroscopy (SE(R)RS) measurements, dynamic light scattering (DLS), and zeta potential analyzes. Compared to manual methods, our automated approach demonstrated higher reproducibility in both synthesis and the properties of the produced nanotags. Additionally, to underscore their potential in sensing applications, we functionalized the nanotags with streptavidin. By combining precise control over nanotags synthesis with robust characterization, this work establishes a new standard for the rapid and reliable production of advanced nanoplatforms for biomedical applications.
Collapse
Affiliation(s)
- Ngoc Nhu Vu
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| | - Ka Wai Ng
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| | - Siddhant Jaitpal
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| | - Masoud Negahdary
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| | - Tran Nguyen
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| | - Rohit Sai Kodam
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| | - Samuel Mabbott
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| |
Collapse
|
148
|
Rogalewicz B, Czylkowska A. Recent advances in the discovery of copper(II) complexes as potential anticancer drugs. Eur J Med Chem 2025; 292:117702. [PMID: 40328033 DOI: 10.1016/j.ejmech.2025.117702] [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: 02/13/2025] [Revised: 04/13/2025] [Accepted: 04/27/2025] [Indexed: 05/08/2025]
Abstract
This review article offers a literature search of the most active, new copper (II) anticancer complexes based on nitrogen-containing ligands, reported in the literature over the past 5 years: from the beginning of 2019, until mid-2024. In the modern world, cancer remains one of the deadliest diseases of all. Although years of the ongoing research allowed us to better understand its nature, and thus aim more precisely at specific molecular targets and pathways, many of its aspects remain unclear. Today, chemotherapy still remains at the forefront of cancer treatment. With the ever-growing struggles to overcome chemoresistance and occurrence of serious side effects, the discovery of new, more selective and active drugs is a task of an utmost importance. At the same time, copper (II)-based compounds offer a wide array of biological activities and valuable biochemical properties. This review article provides the update on the recent advances in the discovery of new potential anticancer drugs among copper (II)-based compounds in the recent five years.
Collapse
Affiliation(s)
- Bartłomiej Rogalewicz
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924, Lodz, Poland.
| | - Agnieszka Czylkowska
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924, Lodz, Poland.
| |
Collapse
|
149
|
Kong X, Lei L, Ma Z. LINC00704 facilitates cell proliferation, migration, and invasion via miR-323a-3p/SLC44A1 axis in epithelial ovarian cancer. Discov Oncol 2025; 16:640. [PMID: 40301147 PMCID: PMC12040790 DOI: 10.1007/s12672-025-01866-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 02/03/2025] [Indexed: 05/01/2025] Open
Abstract
Epithelial ovarian cancer (EOC) is the most common subtype of ovarian cancer and is a highly recurrent and lethal malignancy of the female reproductive system. Accumulating evidence has indicated that long non-coding RNAs (lncRNAs) play pivotal roles in tumorigenesis. Long intergenic non-protein coding RNA 704 (LINC00704) has been recognized as an oncogenic lncRNA in several malignancies. However, its specific role in EOC remains unclear. In this study, RT-qPCR revealed the upregulation of LINC00704 in EOC tissues and cell lines. CCK-8 and EdU assays showed that LINC00704 knockdown inhibited EOC cell proliferation. Flow cytometry analysis demonstrated that LINC00704 silencing induced EOC cell cycle arrest and apoptosis. Transwell assays indicated the inhibitory effects of LINC00704 silencing on EOC cell migration and invasion. Mechanistically, bioinformatics analysis, RNA pull-down, luciferase reporter and RNA immunoprecipitation assays revealed that LINC00704 upregulated SLC44A1 expression by competitively binding to miR-323a-3p. Moreover, rescue experiments showed that SLC44A1 overexpression could reverse LINC00704 silencing-mediated effects on the malignant behaviors of EOC cells. In conclusion, LINC00704 promotes EOC cell aggressiveness in vitro by regulating the miR-323a-3p/SLC44A1 axis.
Collapse
Affiliation(s)
- Xiaojuan Kong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Hunan University of Chinese Medicine, 95 Shaoshan Middle Road, Yuhua District, Changsha City, Hunan Province, China
| | - Lei Lei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Hunan University of Chinese Medicine, 95 Shaoshan Middle Road, Yuhua District, Changsha City, Hunan Province, China.
| | - Zhengjiao Ma
- Department of Obstetrics and Gynecology, Wancheng District Traditional Chinese Medicine Hospital in Nanyang City, Nanyang, 473000, China
| |
Collapse
|
150
|
Gao C, Li J, Shan B. Research progress on the regulatory role of lactate and lactylation in tumor microenvironment. Biochim Biophys Acta Rev Cancer 2025:189339. [PMID: 40311713 DOI: 10.1016/j.bbcan.2025.189339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
The tumor microenvironment (TME) arises from the dynamic interactions between tumor cells and the surrounding medium, including a variety of cell types and extracellular components, which have an important impact on the genesis and development of tumors. A key player in TME is lactate, a metabolic byproduct of glycolysis, which serves as a significant energy source. Lactate has direct implications on the survival and differentiation of immune cells, the metabolic reprogramming and progression of tumor cells. Moreover, lactylation, a unique post-translational modification, exerts a regulatory effect on TME by affecting gene transcription via adding lactate groups to both histone and non-histone proteins. This review systematically and comprehensively synthesizes emerging evidence on how the lactate-lactylation axis drives immune evasion, therapy resistance, and TME remodeling, highlighting the therapeutic targets related to lactate and lactylation that dismantle this metabolic-epigenetic crosstalk.
Collapse
Affiliation(s)
- Chunyan Gao
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Jiali Li
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Baoen Shan
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China.
| |
Collapse
|