1
|
Noor RAM, Shah NSM, Zin AAM, Sulaiman WAW. Novel cytochrome c-oxidase deficiency and mitochondrial myopathy in non-syndromic cleft lip with or without palate. Tissue Cell 2025; 95:102887. [PMID: 40187006 DOI: 10.1016/j.tice.2025.102887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/22/2025] [Accepted: 03/22/2025] [Indexed: 04/07/2025]
Abstract
The role of mitochondria in regulating cells during the embryonic phase is crucial, yet their function in causing deformation to orbicularis oris muscle of cleft lip remains controversial. The study was performed to explore morphological abnormalities of orbicularis oris muscle and mitochondria in patients with cleft lip with or without palate. Seventeen cleft lip tissue samples were obtained from consented patients who underwent cleft lip repair. All tissue samples were processed according to the respective analyses: modified Gomori trichrome, cytochrome c-oxidase (COX), adenosine triphosphatase (ATP-ase), and transmission electron microscopy (TEM). Fiber types and the sizes of fibers and mitochondria were determined using ImageJ processing program. The pathological hallmark of mitochondrial myopathy, which was the presence of ragged red fibers, and the presence of novel COX-negative fibers were observed in cleft lip tissues. Muscle fiber type ratio for type II fibers was found to be predominant in orbicularis oris muscle of cleft lip tissues with 78.43 %, while type I fibers was 21.57 %. The diameters of the fibers were 6.37 µm for type I, and 6.87 µm for type II. By means of electron microscopy, minimal accumulation of mitochondria and excessive lipid droplets were noted within the myofibrils of cleft lip orbicularis oris muscle. This report on series of pathologic features of the orbicularis oris muscle and mitochondria provides insights into the possibility of impaired mitochondrial function in non-syndromic cleft lip.
Collapse
Affiliation(s)
- Rabiatul Adawiyah Mohamad Noor
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian, Kelantan 16150, Malaysia
| | - Nurul Syazana Mohamad Shah
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian, Kelantan 16150, Malaysia.
| | - Anani Aila Mat Zin
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian, Kelantan 16150, Malaysia
| | - Wan Azman Wan Sulaiman
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian, Kelantan 16150, Malaysia
| |
Collapse
|
2
|
Wang J, Shangguan Y, Long F, Guo Y, Wang H, Chen L. Embryonic exposure to prednisone induces bone developmental toxicity in zebrafish: Characteristics and molecular mechanisms. JOURNAL OF HAZARDOUS MATERIALS 2025; 491:137996. [PMID: 40122004 DOI: 10.1016/j.jhazmat.2025.137996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
As a synthetic glucocorticoid, prednisone has been widely used in autoimmune diseases, recurrent abortion and asthma during pregnancy. Although studies suggested that glucocorticoid exposure during pregnancy have developmental toxicity, systematic research on the characteristics of the developmental toxicity of prednisone is lacking. This study intends to construct embryonic prednisone exposure (EPE) model to observe its bone developmental toxicity characteristics of prednisone and explore the mechanism. The results showed that EPE can shortened body and head length, reduced eye and head area, decreased operculum mineralization area, reduced mineralized vertebrae number, shortened ceratohyal and palatoquadrate cartilage length, and decreased expression of key osteogenic differentiation and cartilage development genes. The toxicity to osteogenesis is more severe than chondrogenesis. The toxicity caused by exposure in the middle and terminal stages of embryogenesis is more serious and shows a concentration-effect relationship. We confirmed that Gr/Hdac6 signaling activation mediates prednisone-induced inhibition of osteoblast differentiation by epigenetically regulating the Postnb/Wnt/β-catenin signaling pathway. The results of this study systematically demonstrate the characteristics of prednisone-induced systemic, bone, and cartilage developmental toxicity, and clarify the epigenetic mechanism of its osteogenic developmental toxicity. This provides theoretical and experimental evidence for the safe use of prednisone during pregnancy and the determination of early monitoring targets for bone developmental toxicity.
Collapse
Affiliation(s)
- Jiaqi Wang
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Yangfan Shangguan
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Fei Long
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Yu Guo
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
3
|
Farinella R, Felici A, Peduzzi G, Testoni SGG, Costello E, Aretini P, Blazquez-Encinas R, Oz E, Pastore A, Tacelli M, Otlu B, Campa D, Gentiluomo M. From classical approaches to artificial intelligence, old and new tools for PDAC risk stratification and prediction. Semin Cancer Biol 2025; 112:71-92. [PMID: 40147701 DOI: 10.1016/j.semcancer.2025.03.004] [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: 11/28/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is recognized as one of the most lethal malignancies, characterized by late-stage diagnosis and limited therapeutic options. Risk stratification has traditionally been performed using epidemiological studies and genetic analyses, through which key risk factors, including smoking, diabetes, chronic pancreatitis, and inherited predispositions, have been identified. However, the multifactorial nature of PDAC has often been insufficiently addressed by these methods, leading to limited precision in individualized risk assessments. Advances in artificial intelligence (AI) have been proposed as a transformative approach, allowing the integration of diverse datasets-spanning genetic, clinical, lifestyle, and imaging data into dynamic models capable of uncovering novel interactions and risk profiles. In this review, the evolution of PDAC risk stratification is explored, with classical epidemiological frameworks compared to AI-driven methodologies. Genetic insights, including genome-wide association studies and polygenic risk scores, are discussed, alongside AI models such as machine learning, radiomics, and deep learning. Strengths and limitations of these approaches are evaluated, with challenges in clinical translation, such as data scarcity, model interpretability, and external validation, addressed. Finally, future directions are proposed for combining classical and AI-driven methodologies to develop scalable, personalized predictive tools for PDAC, with the goal of improving early detection and patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Sabrina Gloria Giulia Testoni
- Division of Gastroenterology and Gastrointestinal Endoscopy, IRCCS Policlinico San Donato, Vita-Salute San Raffaele University, Milan, Italy
| | - Eithne Costello
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Paolo Aretini
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Ricardo Blazquez-Encinas
- Department of Cell Biology, Physiology and Immunology, University of Cordoba / Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Elif Oz
- Department of Biostatistics and Bioinformatics, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Aldo Pastore
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Matteo Tacelli
- Pancreas Translational & Clinical Research Center, Pancreato-Biliary Endoscopy and Endosonography Division, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Burçak Otlu
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, Ankara, Turkey
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | | |
Collapse
|
4
|
Tacelli M, Gentiluomo M, Biamonte P, Castano JP, Berković MC, Cives M, Kapitanović S, Marinoni I, Marinovic S, Nikas I, Nosáková L, Pedraza-Arevalo S, Pellè E, Perren A, Strosberg J, Campa D, Capurso G. Pancreatic neuroendocrine neoplasms (pNENs): Genetic and environmental biomarkers for risk of occurrence and prognosis. Semin Cancer Biol 2025; 112:112-125. [PMID: 40158764 DOI: 10.1016/j.semcancer.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Pancreatic neuroendocrine neoplasms (pNENs) are rare and heterogeneous tumors arising from neuroendocrine cells, representing approximately 10 % of all Gastro-Entero-Pancreatic neuroendocrine neoplasms. While most pNENs are sporadic, a subset is associated with genetic syndromes such as multiple endocrine neoplasia type 1 (MEN1) or von Hippel-Lindau disease (VHL). pNENs are further classified into functioning and non-functioning tumors, with distinct clinical behaviors, prognoses, and treatment approaches. This review explores genetic and environmental biomarkers that influence the risk, prognosis, and therapeutic responses in pNENs. The epidemiology of pNENs reveals an increasing incidence, primarily due to advancements in imaging techniques. Genetic factors play a pivotal role, with germline mutations in MEN1, VHL, and other genes contributing to familial pNENs. Somatic mutations, including alterations in the mTOR pathway and DNA maintenance genes such as DAXX and ATRX, are critical in sporadic pNENs. These mutations, along with epigenetic dysregulation and transcriptomic alterations, underpin the diverse clinical and molecular phenotypes of pNENs. Emerging evidence suggests that epigenetic changes, including DNA methylation profiles, can stratify pNEN subtypes and predict disease progression. Environmental and lifestyle factors, such as diabetes, smoking, and chronic pancreatitis, have been linked to an increased risk of sporadic pNENs. While the association between these factors and tumor progression is still under investigation, their potential role in influencing therapeutic outcomes warrants further study. Advances in systemic therapies, including somatostatin analogs, mTOR inhibitors, and tyrosine kinase inhibitors, have improved disease management. Biomarkers such as Ki-67, somatostatin receptor expression, and O6-methylguanine-DNA methyltransferase (MGMT) status are being evaluated for their predictive value. Novel approaches, including the use of circulating biomarkers (NETest, circulating tumor cells, and ctDNA) and polygenic risk scores, offer promising avenues for non-invasive diagnosis and monitoring. Despite these advancements, challenges remain, including the need for large, well-annotated datasets and validated biomarkers. Future research should integrate multi-omics approaches and leverage liquid biopsy technologies to refine diagnostic, prognostic, and therapeutic strategies. Interdisciplinary collaborations and global consortia are crucial for overcoming current limitations and translating research findings into clinical practice. These insights hold promise for improving prevention, early detection, and tailored treatments, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Matteo Tacelli
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Paolo Biamonte
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Justo P Castano
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Reina Sofia University Hospital, Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Maja Cigrovski Berković
- Department for Sport and Exercise Medicine, Faculty of Kinesiology University of Zagreb, Zagreb 10000, Croatia
| | - Mauro Cives
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy; Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Sanja Kapitanović
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Ilaria Marinoni
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Sonja Marinovic
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Ilias Nikas
- Medical School, University of Cyprus, Nicosia, Cyprus
| | - Lenka Nosáková
- Clinic of Internal Medicine - Gastroenterology, JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Bratislava, Slovakia
| | - Sergio Pedraza-Arevalo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Reina Sofia University Hospital, Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Eleonora Pellè
- Department of GI Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Aurel Perren
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Jonathan Strosberg
- Department of GI Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
5
|
Daniel N, Farinella R, Belluomini F, Fajkic A, Rizzato C, Souček P, Campa D, Hughes DJ. The relationship of the microbiome, associated metabolites and the gut barrier with pancreatic cancer. Semin Cancer Biol 2025; 112:43-57. [PMID: 40154652 DOI: 10.1016/j.semcancer.2025.03.002] [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: 11/30/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Pancreatic cancers have high mortality and rising incidence rates which may be related to unhealthy western-type dietary and lifestyle patterns as well as increasing body weights and obesity rates. Recent data also suggest a role for the gut microbiome in the development of pancreatic cancer. Here, we review the experimental and observational evidence for the roles of the oral, gut and intratumoural microbiomes, impaired gut barrier function and exposure to inflammatory compounds as well as metabolic dysfunction as contributors to pancreatic disease with a focus on pancreatic ductal adenocarcinoma (PDAC) initiation and progression. We also highlight some emerging gut microbiome editing techniques currently being investigated in the context of pancreatic disease. Notably, while the gut microbiome is significantly altered in PDAC and its precursor diseases, its utility as a diagnostic and prognostic tool is hindered by a lack of reproducibility and the potential for reverse causality in case-control cohorts. Future research should emphasise longitudinal and mechanistic studies as well as integrating lifestyle exposure and multi-omics data to unravel complex host-microbiome interactions. This will allow for deeper aetiologic and mechanistic insights that can inform treatments and guide public health recommendations.
Collapse
Affiliation(s)
- Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | | | | | - Almir Fajkic
- Department of Pathophysiology Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Pavel Souček
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic; Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland.
| |
Collapse
|
6
|
Murray K, Oldfield L, Stefanova I, Gentiluomo M, Aretini P, O'Sullivan R, Greenhalf W, Paiella S, Aoki MN, Pastore A, Birch-Ford J, Rao BH, Uysal-Onganer P, Walsh CM, Hanna GB, Narang J, Sharma P, Campa D, Rizzato C, Turtoi A, Sever EA, Felici A, Sucularli C, Peduzzi G, Öz E, Sezerman OU, Van der Meer R, Thompson N, Costello E. Biomarkers, omics and artificial intelligence for early detection of pancreatic cancer. Semin Cancer Biol 2025; 111:76-88. [PMID: 39986585 DOI: 10.1016/j.semcancer.2025.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is frequently diagnosed in its late stages when treatment options are limited. Unlike other common cancers, there are no population-wide screening programmes for PDAC. Thus, early disease detection, although urgently needed, remains elusive. Individuals in certain high-risk groups are, however, offered screening or surveillance. Here we explore advances in understanding high-risk groups for PDAC and efforts to implement biomarker-driven detection of PDAC in these groups. We review current approaches to early detection biomarker development and the use of artificial intelligence as applied to electronic health records (EHRs) and social media. Finally, we address the cost-effectiveness of applying biomarker strategies for early detection of PDAC.
Collapse
Affiliation(s)
- Kate Murray
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Lucy Oldfield
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Irena Stefanova
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | | | | | - Rachel O'Sullivan
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - William Greenhalf
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Salvatore Paiella
- Pancreatic Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Italy
| | - Mateus N Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Brazil
| | - Aldo Pastore
- Fondazione Pisana per la Scienza, Scuola Normale Superiore di Pisa, Italy
| | - James Birch-Ford
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Bhavana Hemantha Rao
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Czech Republic
| | - Pinar Uysal-Onganer
- School of Life Sciences, Cancer Mechanisms and Biomarkers Group, The University of Westminster, United Kingdom
| | - Caoimhe M Walsh
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - George B Hanna
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | | | | | | | | | - Andrei Turtoi
- Tumor Microenvironment and Resistance to Treatment Lab, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, France
| | - Elif Arik Sever
- Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | | | | | | | - Elif Öz
- Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | - Osman Uğur Sezerman
- Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | | | | | - Eithne Costello
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom.
| |
Collapse
|
7
|
Yuan Y, Chen L. Transporters in vitamin uptake and cellular metabolism: impacts on health and disease. LIFE METABOLISM 2025; 4:loaf008. [PMID: 40444179 PMCID: PMC12121362 DOI: 10.1093/lifemeta/loaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/20/2025] [Accepted: 03/01/2025] [Indexed: 06/02/2025]
Abstract
Vitamins are vital nutrients essential for metabolism, functioning as coenzymes, antioxidants, and regulators of gene expression. Their absorption and metabolism rely on specialized transport proteins that ensure bioavailability and cellular utilization. Water-soluble vitamins, including B-complex and vitamin C, are transported by solute carrier (SLC) family proteins and ATP-binding cassette (ABC) transporters for efficient uptake and cellular distribution. Fat-soluble vitamins (A, D, E, and K) rely on lipid-mediated pathways through proteins like scavenger receptor class B type I (SR-BI), CD36, and Niemann-Pick C1-like 1 (NPC1L1), integrating their absorption with lipid metabolism. Defective vitamin transporters are associated with diverse metabolic disorders, including neurological, hematological, and mitochondrial diseases. Advances in structural and functional studies of vitamin transporters highlight their tissue-specific roles and regulatory mechanisms, shedding light on their impact on health and disease. This review emphasizes the significance of vitamin transporters and their potential as therapeutic targets for deficiencies and related chronic conditions.
Collapse
Affiliation(s)
- Yaxuan Yuan
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer, Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical sciences, Zhengzhou University, Zhengzhou, Henan, China, 450001
| | - Ligong Chen
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer, Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical sciences, Zhengzhou University, Zhengzhou, Henan, China, 450001
| |
Collapse
|
8
|
Fang Y, Tan C, Zheng Z, Yang J, Tang J, Guo R, Silli EK, Chen Z, Chen J, Ge R, Liu Y, Wen X, Liang J, Zhu Y, Jin Y, Li Q, Wang Y. The function of microRNA related to cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. Biochem Pharmacol 2025; 236:116849. [PMID: 40056941 DOI: 10.1016/j.bcp.2025.116849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignant tumor characterized by a poor prognosis. A prominent feature of PDAC is the rich and dense stroma present in the tumor microenvironment (TME), which significantly hinders drug penetration. Cancer-associated fibroblasts (CAFs), activated fibroblasts originating from various cell sources, including pancreatic stellate cells (PSCs) and mesenchymal stem cells (MSCs), play a critical role in PDAC progression and TME formation. MicroRNAs (miRNAs) are small, single-stranded non-coding RNA molecules that are frequently involved in tumorigenesis and progression, exhibiting either oncolytic or oncogenic activity. Increasing evidence suggests that aberrant expression of miRNAs can mediate interactions between cancer cells and CAFs, thereby providing novel therapeutic targets for PDAC treatment. In this review, we will focus on the potential roles of miRNAs that target CAFs or CAFs-derived exosomes in PDAC progression, highlighting the feasibility of therapeutic strategies aimed at restoring aberrantly expressed miRNAs associated with CAFs, offering new pathways for the clinical management of PDAC.
Collapse
Affiliation(s)
- Yaohui Fang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Chunlu Tan
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenjiang Zheng
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jiali Tang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruizhe Guo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Epiphane K Silli
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Zhe Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jia Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruyu Ge
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yuquan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xiuqi Wen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jingdan Liang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yunfei Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yutong Jin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Qian Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ying Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
9
|
Fayazi M, Rostami M, Amiri Moghaddam M, Nasiri K, Tadayonfard A, Roudsari MB, Ahmad HM, Parhizgar Z, Majbouri Yazdi A. A state-of-the-art review of the recent advances in drug delivery systems for different therapeutic agents in periodontitis. J Drug Target 2025; 33:612-647. [PMID: 39698877 DOI: 10.1080/1061186x.2024.2445051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
Periodontitis (PD) is a chronic gum illness that may be hard to cure for a number of reasons, including the fact that no one knows what causes it, the side effects of anti-microbial treatment, and how various kinds of bacteria interact with one another. As a result, novel therapeutic approaches for PD treatment must be developed. Additionally, supplementary antibacterial regimens, including local and systemic medication administration of chemical agents, are necessary for deep pockets to assist with mechanical debridement of tooth surfaces. As our knowledge of periodontal disease and drug delivery systems (DDSs) grows, new targeted delivery systems like extracellular vesicles, lipid-based nanoparticles (NPs), metallic NPs, and polymer NPs have been developed. These systems aim to improve the targeting and precision of PD treatments while reducing the systemic side effects of antibiotics. Nanozymes, photodermal therapy, antibacterial metallic NPs, and traditional PD therapies have all been reviewed in this research. Medicinal herbs, antibiotics, photothermal therapy, nanozymes, antibacterial metallic NPs, and conventional therapies for PD have all been examined in this research. After that, we reviewed the key features of many innovative DDSs and how they worked for PD therapy. Finally, we have discussed the advantages and disadvantages of these DDSs.
Collapse
Affiliation(s)
- Mehrnaz Fayazi
- School of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Mitra Rostami
- School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Kamyar Nasiri
- Department of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Azadeh Tadayonfard
- Department of Prosthodontics, Dental Faculty, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Behnam Roudsari
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Zahra Parhizgar
- Resident of Periodontology, Department of Periodontics, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
10
|
Göktaş M, Karabulut D, Ünlü A, Achmet G, Tunçbilek N. MRI-based radiogenomics analysis for predicting prognosis and XRCC1 gene polymorphism in pancreatic ductal adenocarcinoma. Clin Transl Oncol 2025; 27:2517-2526. [PMID: 39487951 DOI: 10.1007/s12094-024-03763-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 10/10/2024] [Indexed: 11/04/2024]
Abstract
PURPOSE To evaluate the prognostic effects of apparent diffusion coefficient (ADC) values, qualitative MRI findings, and XRCC1 polymorphism in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS Between January 2019 and December 2021, 41 PDAC patients (23 males; 66.6 ± 8.9 years) diagnosed with MRI and treated with chemotherapy were included in this prospective, unicenter study. Quantitative b:0-800 ADC values were calculated at the workstation using a circular region of interest with a diameter of 2 cm2. Polymerase chain reaction restriction fragment length polymorphism was used to detect XRCC1 genotypes from blood samples. Demographic data, MRI findings, and survival times were recorded. Sensitivity, specificity of ADCmin values, and relationship between XRCC1 genes in predicting survival were calculated and compared. RESULTS The median overall survival time was calculated as 9.27 ± 1.4 months. The cut-off value of ADCmin was found to be 0.996 × 10-3 mm2/s for predicting a 4.6-month survival with 77.3% sensitivity and 84.2% specificity. The distribution of XRCC1 codon 399 polymorphism was determined as 26.8% (n = 11) GG, 65.9% (n = 27) AG, and 7.3% (n = 3) AA. There was no statistical correlation between ADCmin values and XRCC1 gene polymorphism (p > 0.05). ADCmin < 0.996 × 10-3 and the XRCC1 codon 399 AG/AA genotype was the subgroup with the worst prognosis (p = 0.035). The mean age at diagnosis was 71.0 ± 9.1 years in cases with GG genotype, while it was 64.6 ± 8.9 years in cases with AG/AA genotype, which was statistically significant (p = 0.038). CONCLUSIONS ADCmin values are useful for predicting prognosis in patients with PDAC. XRCC1 gene polymorphism may affect the age at diagnosis.
Collapse
Affiliation(s)
- Muhammet Göktaş
- Department of Radiology, Trakya University, Edirne, Turkey.
- Department of Radiology, Çerkezköy State Hospital, Tekirdağ, Turkey.
| | | | - Ayhan Ünlü
- Department of Biophysics, Trakya University, Edirne, Turkey
| | | | | |
Collapse
|
11
|
Mohammadi Y, Babaeenezhad E, Yarahmadi S. MiRNA-211 Dysregulation contributes to the pathogenesis of digestive tract cancers: Mechanistic and clinical implications. Gene 2025; 962:149596. [PMID: 40414469 DOI: 10.1016/j.gene.2025.149596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/25/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Gastrointestinal tract cancers are a significant cause of mortality, with cancers of the colon, stomach, liver, and pancreas being the most common types of cancer. Early detection is crucial for improving survival rates. Epigenetic modifications, such as microRNA (miRNA) gene silencing, play a crucial role in the development and progression of gastrointestinal cancer. Aberrant miRNA expression may contribute to cancer growth, proliferation, apoptosis, and drug resistance. This study focuses on miRNA-211, a dysregulated miRNA in gastrointestinal cancer that may act as a tumor suppressor or promoter. The research investigates the upstream regulatory elements and target genes of miRNA-211 to reveal its potential as a diagnostic and therapeutic marker for digestive tract malignancies.
Collapse
Affiliation(s)
- Yaser Mohammadi
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran; Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Esmaeel Babaeenezhad
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran; Department of Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Sahar Yarahmadi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran; Department of Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
12
|
Hu D, Xu B, Huang G, Hu X, Li J, Chen Z, Liu W, Wen Z. CALB2 facilitates macrophage M2 polarization to promote the growth and metastasis of pancreatic adenocarcinoma. Cell Signal 2025; 134:111887. [PMID: 40409389 DOI: 10.1016/j.cellsig.2025.111887] [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: 02/14/2025] [Revised: 05/02/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
Tumor-associated macrophages mainly differentiate into M2 phenotypes, which secrete cytokines that reshape the tumor microenvironment and promote tumor progression. This study was to explore the mechanism of CALB2 in M2 polarization and pancreatic adenocarcinoma (PAAD). Clinical tissue samples of PAAD were collected, followed by detection of WTAP, FOSL1, and CALB2 expression. The correlation between WTAP and FOSL1 or between FOSL1 and CALB2 was analyzed. THP1 cells were induced into M0 macrophages, followed by plasmid transfection and induction of M2-type macrophages. After macrophages were co-cultured with PAAD cells, functional experiments were designed to evaluate PAAD cell malignant behaviors. A transplantation tumor model and a liver metastasis model were established to assess tumor growth and metastasis. High expression of WTAP, FOSL1, and CALB2 was found in PAAD tissues and M2-type macrophages. WTAP positively linked with FOSL1, so as FOSL1 and CALB2. Mechanistically, WTAP enhanced m6A modification of FOSL1 to promote its expression, and FOSL1 promoted CALB2 transcription. Knockdown of WTAP, FOSL1, or CALB2 in macrophages inhibited PAAD cell malignant behaviors, which could be reversed by CALB2 upregulation. WTAP knockdown restrained the growth and metastasis of PAAD in nude mice via the FOSL1/CALB2 axis. In conclusion, WTAP increased the m6A level of FOSL1, activated CALB2 transcription, and promoted M2 polarization of macrophages, thereby promoting the growth and metastasis of PAAD.
Collapse
Affiliation(s)
- Dongwei Hu
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang 325000, PR China
| | - Bo Xu
- Department of Hepato-pancreato-biliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Guoyu Huang
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, AZ 85259, USA
| | - Xiaowei Hu
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang 325000, PR China
| | - Jinjie Li
- Department of Hepato-pancreato-biliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Zongjing Chen
- Department of Hepato-pancreato-biliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Wei Liu
- Institute of Digestive Disease, China Three Gorges University, Yichang, PR China.
| | - Zhengde Wen
- Department of Hepato-pancreato-biliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China; Wenzhou Key Laboratory of Perioperative Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China.
| |
Collapse
|
13
|
Ding Z, Yang L, Wang X, Wang Y, Chen X. The expression and prognostic value of CCL19 in breast cancer. Discov Oncol 2025; 16:830. [PMID: 40392467 PMCID: PMC12092893 DOI: 10.1007/s12672-025-02715-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 05/14/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Breast cancer ranks as the foremost cause of cancer-related mortality among women globally. Timely diagnosis stands as the most effective approach in mitigating breast cancer mortality rates. There exists a close relationship between immune processes and tumorigenesis. This study aims to elucidate the immune mechanisms and potential biomarkers associated with breast cancer using bioinformatics techniques. OBJECTIVE Initially, differentially expressed genes were identified through consensus analysis of invasive breast cancer (BRCA) samples sourced from The Cancer Genome Atlas (TCGA) database, focusing on immunotherapy response. Subsequently, protein-protein interaction (PPI) networks, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were employed to refine the selection of potential genes. Lastly, expression and prognostic analyses of hub genes were conducted to identify reliable key genes, with a focus on CCL19. Immunohistochemistry (IHC) was employed to assess the differential expression of CCL19 in both tumor and adjacent breast tissue samples. Additionally, protein correlation analysis, signaling pathway analysis, immune infiltration analysis, gene co-expression analysis, and drug sensitivity analysis of CCL19 were conducted to investigate its pathological clinical features and potential biological functions. RESULTS CCL19 expression exhibited a significant increase in breast cancer. Elevated CCL19 expression correlates with advanced tumor stage and indicates a favorable prognosis in breast cancer. CCL19 expression correlates with the abundance of diverse tumor-infiltrating immune cells (TIICs). CCL19 exhibits a positive correlation with the expression of the majority of immune-related genes. Enrichment analysis revealed the involvement of CCL19 in immune-related pathways and the PI3K-Akt signaling pathway. These findings suggest that CCL19 may influence breast cancer prognosis through immune infiltration. Patients exhibiting high CCL19 expression demonstrated more favorable responses to immunotherapy. CONCLUSION Breast cancer demonstrates overexpression of CCL19. CCL19 holds promise as a biomarker for forecasting breast cancer prognosis and as a potential therapeutic target.
Collapse
Affiliation(s)
- Zonghao Ding
- General Surgery Department, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Yang
- AnHui Medical University, Hefei, China
| | | | - Yong Wang
- General Surgery Department, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiao Chen
- Thyroid and Breast Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
14
|
Selvi S, Real CM, Gentiluomo M, Balounova K, Vokacova K, Cumova A, Mohlenikova-Duchonova B, Rizzato C, Halasova E, Vodickova L, Smolkova B, Hemminki K, Campa D, Vodicka P. Genomic instability, DNA damage response and telomere homeostasis in pancreatic cancer. Semin Cancer Biol 2025; 113:59-73. [PMID: 40378535 DOI: 10.1016/j.semcancer.2025.05.005] [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: 04/16/2025] [Accepted: 05/04/2025] [Indexed: 05/19/2025]
Abstract
Pancreatic cancer (PC) is becoming one of the most serious health problems at present, but its causes and risk factors are still unclear. One of the drivers in pancreatic carcinogenesis is altered genomic (DNA) integrity with subsequent genomic instability in cancer cells. The latter comprises a) DNA damage response and DNA repair mechanisms, b) DNA replication and mitosis, c) epigenetic regulation, and d) telomere maintenance. In our review we addressed the above aspects in relation to the most abundant and severe form of PC, pancreatic ductal adenocarcinoma (PDAC). In summary, the interactions between the DNA damage response, telomere homeostasis and mitotic regulation are not comprehensively understood at present, including the epigenetic factors entering the trait of genomic stability maintenance. In addition, the complexity of telomere homeostasis in relation to PDAC risk, prognosis and prediction also warrants further investigations.
Collapse
Affiliation(s)
- Saba Selvi
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic; Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague 12800, Czech Republic
| | - Carmen Macías Real
- Cancer Predisposition and Biomarkers Group, Instituto de Investigacion Sanitaria de Santiago, Santiago de Compostela, Spain
| | | | - Katerina Balounova
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic; Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Klara Vokacova
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Andrea Cumova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava 84505, Slovakia
| | | | - Cosmeri Rizzato
- Department of Biology, University of Pisa, Pisa 56123, Italy
| | - Erika Halasova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4, Martin 03601, Slovakia
| | - Ludmila Vodickova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4, Martin 03601, Slovakia; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, Pilsen 32300, Czech Republic; Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, Prague 12800, Czech Republic
| | - Bozena Smolkova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava 84505, Slovakia
| | - Kari Hemminki
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, Pilsen 32300, Czech Republic; Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, FRG 69120, Germany
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa 56123, Italy
| | - Pavel Vodicka
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4, Martin 03601, Slovakia; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, Pilsen 32300, Czech Republic; Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, Prague 12800, Czech Republic.
| |
Collapse
|
15
|
Roscigno G, Jacobs S, Toledo B, Borea R, Russo G, Pepe F, Serrano MJ, Calabrò V, Troncone G, Giovannoni R, Giovannetti E, Malapelle U. The potential application of stroma modulation in targeting tumor cells: focus on pancreatic cancer and breast cancer models. Semin Cancer Biol 2025:S1044-579X(25)00060-4. [PMID: 40373890 DOI: 10.1016/j.semcancer.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and spreading being considered as "the dark side of the tumor". Within this term tumor cells, immune components, supporting cells, extracellular matrix and a myriad of bioactive molecules that synergistically promote tumor development and therapeutic resistance, are included. Recent findings revealed the profound impacts of TME on cancer development, serving as physical support, critical mediator and biodynamic matrix in cancer evolution, immune modulation, and treatment outcomes. TME targeting strategies built on vasculature, immune checkpoints, and immuno-cell therapies, have paved the way for revolutionary clinical interventions. On this basis, the relevance of pre-clinical and clinical investigations has rapidly become fundamental for implementing novel therapeutical strategies breaking cell-cell and cell -mediators' interactions between TME components and tumor cells. This review summarizes the key players in the breast and pancreatic TME, elucidating the intricate interactions among cancer cells and their essential role for cancer progression and therapeutic resistance. Different tumors such breast and pancreatic cancer have both different and similar stroma features, that might affect therapeutic strategies. Therefore, this review aims to comprehensively evaluate recent findings for refining breast and pancreatic cancer therapies and improve patient prognoses by exploiting the TME's complexity in the next future.
Collapse
Affiliation(s)
- Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy.
| | - Sacha Jacobs
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| | - Belen Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain.
| | - Roberto Borea
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Maria Jose Serrano
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain.
| | - Viola Calabrò
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Roberto Giovannoni
- Department of Biology, Genetic Unit, University of Pisa, Via Derna 1, 56126 Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy.
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|
16
|
Sena G, Currò G, Vescio G, Ammerata G, Amaddeo A, Rizzuto A. Surgical vs. Conservative Management of Patients with Nonfunctioning Pancreatic Neuroendocrine Tumors Smaller than 2 cm (NF-PANNETs < 2 cm) Systematic Review and Meta-Analysis. Cancers (Basel) 2025; 17:1649. [PMID: 40427145 PMCID: PMC12109734 DOI: 10.3390/cancers17101649] [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: 04/11/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
INTRODUCTION There is no consensus on managing non-functioning pancreatic neuroendocrine tumors smaller than 2 cm (NF-PANNETs < 2 cm). Therefore, their treatment remains controversial. The aim of this study, by literature review and meta-analysis, is to establish the best management of NF-PANNETs < 2 cm based on overall survival (OS) and cancer-specific survival (CSS). MATERIALS AND METHODS An extensive online search was conducted using the MEDLINE, EMBASE, Google Scholar, Scopus, Web of Science, and Cochrane Central databases. All retrospective and prospective studies were included in this study, comparing the outcomes of surgical management vs. conservative management in patients with NF-PANNETs < 2 cm. The pooled odds ratio and 95% CI for survival were calculated. RESULTS Six studies were included in the quantitative analysis, with 2708 patients managed operatively and 985 managed conservatively. A pooled analysis of all the data demonstrated increased OS in patients managed operatively compared with those managed conservatively at five years (OR = 1.77, 95% CI: 0.96 to 2.58; p = 0.002). In contrast, the meta-analysis did not demonstrate increased CSS in patients undergoing surgical resection compared with conservative management (OR = 1.01, 95% CI: -5.25 to 7.27; p = 0.56). Furthermore, analysis demonstrated a high heterogeneity for OS (Q = 43.98, p < 0.001, tau2 = 0.46, I2 = 88.63%) and for CSS (Q = 22.81, p < 0.0001, tau2 = 1.72, I2 = 91.23%). CONCLUSION This systematic review and meta-analysis indicated that surgical management of NF-PANNETs < 2 cm improves overall survival (OS) but does not significantly enhance cancer-specific survival (CSS). There is variability in outcomes among studies, and while surgery may help some patients, the lack of clear CSS benefits and associated risks call for individualized decision-making. Therefore, a conservative approach with active surveillance may be more suitable for low-risk patients.
Collapse
Affiliation(s)
- Giuseppe Sena
- Department of General Surgery, “Renato Dulbecco” Hospital, Viale Europa, 88100 Catanzaro, Italy
| | - Giuseppe Currò
- Department of Health Science, “Magna Graecia” University, Viale Europa, 88100 Catanzaro, Italy
| | - Giuseppina Vescio
- Department of Surgical and Medical Science, “Magna Graecia” University, Viale Europa, 88100 Catanzaro, Italy; (G.V.); (A.A.); (A.R.)
| | - Giorgio Ammerata
- Department of General Surgery, “Di Venere” Hospital, Via Ospedale Di Venere, 1, 70131 Bari, Italy;
| | - Angela Amaddeo
- Department of Surgical and Medical Science, “Magna Graecia” University, Viale Europa, 88100 Catanzaro, Italy; (G.V.); (A.A.); (A.R.)
| | - Antonia Rizzuto
- Department of Surgical and Medical Science, “Magna Graecia” University, Viale Europa, 88100 Catanzaro, Italy; (G.V.); (A.A.); (A.R.)
| |
Collapse
|
17
|
Fabregat-Franco C, Castet F, Castillo G, Salcedo M, Sierra A, López-Valbuena D, Pando E, Tian TV, Macarulla T. Genomic profiling unlocks new treatment opportunities for ampullary carcinoma. ESMO Open 2025; 10:104480. [PMID: 40359709 DOI: 10.1016/j.esmoop.2025.104480] [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: 11/15/2024] [Revised: 01/12/2025] [Accepted: 01/23/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Ampullary carcinoma (AC) is a rare disease with an abysmal prognosis and few treatment options. The molecular landscape and its therapeutic implications remain inadequately understood. This study aims to provide a clinical and genomic characterization of AC and explore opportunities for precision oncology. MATERIALS AND METHODS We carried out a retrospective analysis of clinical and genomic features in patients with AC treated in our institution. Gene mutations were categorized into molecular pathways, and potentially targetable alterations were classified according to the European Society for Medical Oncology (ESMO) Scale for Clinical Actionability of Molecular Targets (ESCAT). Key molecular findings were validated in an external cohort. RESULTS We included 78 patients with a median age of 66 years; 51.6% were women, and most were treated with surgery (81.2%). Histologically, they were classified as pancreaticobiliary (58.3%), intestinal (INT, 33.3%), and mixed (8.3%). The percentages of patients diagnosed at stages I, II, III, and IV disease were 18.8%, 23.4%, 32.8%, and 25.0%, respectively. Of note, the INT subtype was enriched in transforming growth factor-β pathway alterations (25.9% versus 6.1%, P = 0.03). Potentially actionable molecular alterations were found in 52% of the patients. Importantly, KRASWT tumors were enriched in potentially targetable alterations ESCAT I-IIIA both in our cohort (37.2% versus 9.4%, P = 0.006) and external validation cohort (23.0% versus 9.3%, P = 0.01), including 25.6% ERBB2 amplification/mutation, 14.0% homologous recombination deficiency status, and 7.4% microsatellite instability status. Six patients received matched targeted therapies after progression to chemotherapy, with a response rate of 50% and two patients surviving for >1 year. CONCLUSIONS AC patients are enriched in targetable alterations, especially KRASWT tumors, and could particularly benefit from precision oncology-based approaches.
Collapse
Affiliation(s)
- C Fabregat-Franco
- Medical Oncology Department, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - F Castet
- Upper Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain
| | - G Castillo
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - M Salcedo
- Human Pathology Department, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - A Sierra
- Upper Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain
| | - D López-Valbuena
- Upper Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain
| | - E Pando
- Department of HPB and Transplant Surgery, Vall d'Hebron University Hospital, Barcelona, Spain
| | - T V Tian
- Upper Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain
| | - T Macarulla
- Upper Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain.
| |
Collapse
|
18
|
Peduzzi G, Archibugi L, Farinella R, de Leon Pisani RP, Vodickova L, Vodicka P, Kraja B, Sainz J, Bars-Cortina D, Daniel N, Silvestri R, Uysal-Onganer P, Landi S, Dulińska-Litewka J, Comandatore A, Campa D, Hughes DJ, Rizzato C. The exposome and pancreatic cancer, lifestyle and environmental risk factors for PDAC. Semin Cancer Biol 2025; 113:100-129. [PMID: 40368260 DOI: 10.1016/j.semcancer.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/16/2025]
Abstract
Pancreatic cancer (PC), particularly pancreatic ductal adenocarcinoma (PDAC), is a significant global health issue with high mortality rates. PDAC, though only 3 % of cancer diagnoses, causes 7 % of cancer deaths due to its severity and asymptomatic early stages. Risk factors include lifestyle choices, environmental exposures, and genetic predispositions. Conditions like new-onset type 2 diabetes and chronic pancreatitis also contribute significantly. Modifiable risk factors include smoking, alcohol consumption, non-alcoholic fatty pancreatic disease (NAFPD), and obesity. Smoking and heavy alcohol consumption increase PC risk, while NAFPD and obesity, particularly central adiposity, contribute through chronic inflammation and insulin resistance. Refined sugar and sugar-sweetened beverages (SSBs) are also linked to increased PC risk, especially among younger individuals. Hormonal treatments and medications like statins, aspirin, and metformin have mixed results on PC risk, with some showing protective effects. The gut microbiome influences PC through the gut-pancreas axis, with disruptions leading to inflammation and carcinogenesis. Exposure to toxic substances, including heavy metals and chemicals, is associated with increased PC risk. Glycome changes, such as abnormal glycosylation patterns, are significant in PDAC development and offer potential for early diagnosis. Interactions between environmental and genetic factors are crucial in PDAC susceptibility. Genome-wide association studies (GWAS) have identified several single nucleotide polymorphisms (SNPs) linked to PDAC, but gene-environment interactions remain largely unexplored. Future research should focus on polygenic risk scores (PRS) and large-scale studies to better understand these interactions and their impact on PDAC risk.
Collapse
Affiliation(s)
| | - Livia Archibugi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Ruggero Ponz de Leon Pisani
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ludmila Vodickova
- Biomedical Center Martin, Bioinformatic Center, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Slovakia; Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Vodicka
- Biomedical Center Martin, Bioinformatic Center, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Slovakia; Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Bledar Kraja
- University Clinic of Gastrohepatology, University Hospital Center Mother Teresa, Tirana, Albania
| | - Juan Sainz
- Department of Biochemistry and Molecular Biology, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid 28029, Spain; GENYO. Centre for Genomics and Oncological Research. Genomic Oncology department, Granada, Spain; Instituto de Investigación Biosanitaria Ibs.Granada, Granada, Spain
| | - David Bars-Cortina
- Institut Català d'Oncologia (ICO) IDIBELL, Unit of Biomarkers and Susceptibility (UBS), Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Institut Català d'Oncologia (ICO) IDIBELL, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | | - Pinar Uysal-Onganer
- Cancer Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London, UK
| | - Stefano Landi
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | |
Collapse
|
19
|
Kausar N. Machine learning and explainable artificial intelligence reveals the MicroRNAs associated with survival of head and neck squamous cell carcinoma patients. Comput Biol Chem 2025; 118:108503. [PMID: 40378655 DOI: 10.1016/j.compbiolchem.2025.108503] [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: 11/07/2024] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
Dysregulated microRNAs (miRNAs) play a significant role in cancer development and metastasis. In literature, miRNAs have been used for the survival prediction of different types of cancers using AI. Although AI is useful for diagnosis and prognosis prediction of cancer, however, a major criticism of incorporating it into medical fields is that it is essentially a mechanistically uninterpretable opaque "black box", and hence it may not have the required level of accountability, transparency, and reliability in decisions of cancer diagnosis and prognosis for their adoption in clinical settings. Therefore, there is need to develop intelligent models which may explain their prediction so that they may be reliably used by the clinicians. As dysregulated miRNAs are reported to cause cancer metastasis hence, they can play role in survival of patient. Therefore, there is needed to develop ML based techniques which may automatically indicate specific miRNAs involved in survival of patients. In this research, Machine Learning and Explainable AI (XAI) based models have been developed for survival prediction of Head and Neck Squamous Cell Carcinoma (HNSC) patients using miRNA sequences and clinical datasets. miRNAs dataset contains the data of 485 HNSC patients and clinical dataset contains data of 528 patients. The proposed XAI based model explains its prediction by showing the specific miRNA sequences involved in survival of the patients to demonstrate its reliability to be used by clinicians for therapeutic decisions. In this study, it has been shown that explainable ML can provide explicit knowledge of how models make their predictions, which is necessary for increasing the trust and adoption of innovative ML techniques in oncology and healthcare.
Collapse
Affiliation(s)
- Nabeela Kausar
- Department of Software Engineering and Artificial Intelligence, Iqra University, Islamabad, Pakistan.
| |
Collapse
|
20
|
Spencer KR, King GG. MDM2 as a therapeutic target in advanced biliary tract cancers. Oncologist 2025; 30:oyaf094. [PMID: 40421959 PMCID: PMC12107537 DOI: 10.1093/oncolo/oyaf094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 04/08/2025] [Indexed: 05/28/2025] Open
Abstract
Biliary tract cancers (BTCs) are a heterogeneous group of tumors arising from cells in the bile ducts and gallbladder. The 5-year overall survival rate for all BTC stages combined is ~20%, and treatment options for patients with unresectable disease are limited, leaving an unmet clinical need. In recent years, significant efforts have been made to refine and implement targeted therapeutic approaches for patients with BTC. The adoption of early and comprehensive molecular profiling is crucial to identifying patients who may be candidates for effective targeted therapies. Characterization of the molecular landscape of BTCs led to the identification of murine double minute 2 homolog gene (MDM2) amplification across all BTC subtypes. The MDM2 protein is a critical negative regulator of p53 stabilization and activity that is an emerging actionable biomarker in BTCs. There are multiple therapeutic approaches that aim to target MDM2 activity, thereby restoring the intrinsic tumor suppressor function of p53 and halting oncogenesis. However, these have been limited by our evolving understanding of the role of MDM2 in BTC pathogenesis. Here, we offer a review of the current understanding of the role of MDM2 in BTC biology and its therapeutic implications.
Collapse
Affiliation(s)
- Kristen R Spencer
- Department of Medicine at NYU Grossman School of Medicine, NYU Langone Perlmutter Cancer Center, New York, NY 10016, United States
| | - Gentry G King
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA 98109, United States
| |
Collapse
|
21
|
Earl J, Kataki A, Canzian F, Costello E, Campa D, Greenhalf W. Targeting pancreatic cancer screening by identification of pathogenic variants of BRCA2/ BRCA1 in healthy individuals who have no known family history of pancreatic cancer: The arguments for and against. Semin Cancer Biol 2025; 113:1-8. [PMID: 40339999 DOI: 10.1016/j.semcancer.2025.05.001] [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: 02/10/2025] [Revised: 04/15/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
The majority of patients with pancreatic ductal adenocarcinoma (PDAC) are no longer suitable for treatment with curable intent at the time of diagnosis resulting in a 5-year survival of less than 10 %. Imaging of asymptomatic individuals could identify early cancers, but only with a risk of falsely identifying a benign lesion as malignant. Screening of an unselected population would result in far more such false positives than true early cancers. Selection before screening is therefore essential, but there are very few populations at high enough risk to make screening more beneficial than counterproductive. These populations include carriers of specific mutations in BRCA2, and arguably BRCA1, who have a family history of PDAC. These pathogenic mutations all have a predictable effect in making loss of Homologous Recombination Repair (HRR) likely in a carrier's lifetime. In this review the impact of such loss of HRR function on the likelihood of PDAC development will be discussed. Furthermore, it will be discussed whether the identification of a germline pathogenic mutation is sufficient to justify carrier surveillance for the development of the malignancy, or whether the current practice of screening only those carriers with a close relative diagnosed with PDAC is justifiable, as only a proportion of carriers are at high risk. The review will go beyond this to discuss whether there is an essential need to better define and stratify those at high risk, so that only high-risk carriers are put on surveillance.
Collapse
Affiliation(s)
- Julie Earl
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, Carretera Colmenar Km 9,100, Madrid 28034, Spain
| | - Agapi Kataki
- Department of Propaeudeutic Surgery, Breast Unit, Medical School, National and Kapodistrian University of Athens, Greece
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | | | - William Greenhalf
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
22
|
Mao Y, Li Y, Zheng Z, Xu Y, Ke M, He A, Liang F, Zhang K, Wang X, Gao W, Tian R. All-at-once spatial proteome profiling of complex tissue context with single-cell-type resolution by proximity proteomics. Cell Syst 2025:101291. [PMID: 40345200 DOI: 10.1016/j.cels.2025.101291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/01/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025]
Abstract
Spatial proteomics enables in-depth mapping of tissue architectures, mostly achieved by laser microdissection-mass spectrometry (LMD-MS) and antibody-based imaging. However, trade-offs among sampling precision, throughput, and proteome coverage still limit the applicability of these strategies. Here, we propose proximity labeling for spatial proteomics (PSPro) by combining precise antibody-targeted biotinylation and efficient affinity purification for all-at-once cell-type proteome capture with sub-micrometer resolution from single tissue slice. With fine-tuned labeling parameters, PSPro shows reliable performance in benchmarking against flow cytometry- and LMD-based proteomic workflows. We apply PSPro to tumor and spleen slices, enriching thousands of proteins containing known markers from ten cell types. We further incorporate LMD into PSPro to facilitate comparison of cell subpopulations from the same tissue slice, revealing spatial proteome heterogeneity of cancer cells and immune cells in pancreatic tumor. Collectively, PSPro converts the traditional "antibody-epitope" paradigm to an "antibody-cell-type proteome" for spatial biology in a user-friendly manner. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Yiheng Mao
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuan Li
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhendong Zheng
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yanfen Xu
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mi Ke
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - An He
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Fuchao Liang
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Keren Zhang
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xi Wang
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Weina Gao
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ruijun Tian
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
23
|
Chen C, Wang J, Zhu X, Zhang S, Yuan X, Hu J, Liu C, Liu L, Zhang Z, Li J. Lactylation as a metabolic epigenetic modification: Mechanistic insights and regulatory pathways from cells to organs and diseases. Metabolism 2025; 169:156289. [PMID: 40324589 DOI: 10.1016/j.metabol.2025.156289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/20/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
In recent years, lactylation, a novel post-translational modification, has demonstrated a unique role in bridging cellular metabolism and epigenetic regulation. This modification exerts a dual-edged effect in both cancer and non-cancer diseases by dynamically integrating the supply of metabolic substrates and the activity of modifying enzymes: on one hand, it promotes tissue homeostasis and repair through the activation of repair genes; on the other, it exacerbates pathological progression by driving malignant phenotypes. In the field of oncology, lactylation regulates key processes such as metabolic reprogramming, immune evasion, and therapeutic resistance, thereby shaping the heterogeneity of the tumor microenvironment. In non-cancerous diseases, including neurodegeneration and cardiovascular disorders, its aberrant activation can lead to mitochondrial dysfunction, fibrosis, and chronic inflammation. Existing studies have revealed a dynamic regulatory network formed by the cooperation of modifying and demodifying enzymes, and have identified mechanisms such as subcellular localization and RNA metabolism intervention that influence disease progression. Nevertheless, several challenges remain in the field. This article comprehensively summarizes the disease-specific regulatory mechanisms of lactylation, with the aim of providing a theoretical foundation for its targeted therapeutic application.
Collapse
Affiliation(s)
- Cong Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Xueying Zhu
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shan Zhang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xiandun Yuan
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing 100096, China
| | - Jun Hu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Chao Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Lanchun Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Zhenpeng Zhang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| |
Collapse
|
24
|
Powrózek T, Otieno MO, Maffeo D, Frullanti E, Martinez-Useros J. Blood circulating miRNAs as pancreatic cancer biomarkers: An evidence from pooled analysis and bioinformatics study. Int J Biol Macromol 2025; 308:142469. [PMID: 40180095 DOI: 10.1016/j.ijbiomac.2025.142469] [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: 08/08/2024] [Revised: 03/09/2025] [Accepted: 03/22/2025] [Indexed: 04/05/2025]
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers, characterized by a poor prognosis. Currently, there are no screening programs for the early detection of PC, and existing diagnostic methods are primarily limited to high-risk individuals. Biomarkers such as CA19-9 have not significantly improved early diagnosis, making the identification of new potential biomarkers crucial for routine clinical practice. Among the candidate biomarkers, miRNAs have been most extensively studied due to their role in regulating gene expression (either as oncomiRs or tumor suppressor miRNAs) and their potential for minimally invasive analysis through liquid biopsy techniques. This review aims to summarize the current literature on blood-circulating miRNAs and their diagnostic value in PC detection, considering the context of CA19-9 and benign pancreatic diseases. The data from the collected studies were curated through both statistical and bioinformatics analyses to identify the most promising miRNAs with optimal diagnostic accuracy for PC detection and to assess their role in the molecular processes leading to tumor development.
Collapse
Affiliation(s)
- Tomasz Powrózek
- Department of Human Physiology, Medical University of Lublin, Lublin, Poland.
| | - Michael Ochieng' Otieno
- Translational Oncology Division, Oncohealth Institute, Fundacion Jiménez Díaz University Hospital, Madrid, Spain
| | - Debora Maffeo
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Cancer Genomics and Systems Biology Lab, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elisa Frullanti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Cancer Genomics and Systems Biology Lab, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Javier Martinez-Useros
- Translational Oncology Division, Oncohealth Institute, Fundacion Jiménez Díaz University Hospital, Madrid, Spain; Area of Physiology, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos University, Madrid, Spain
| |
Collapse
|
25
|
Barthélémy D, Vigneron A, Rousset X, Guitton J, Grolleau E, Raffin M, Balandier J, Lescuyer G, Bardou M, Geiguer F, Couraud S, Bardel C, Viallet J, Benzerdjeb N, Payen L. Pharmacological effects of osimertinib on a chicken chorioallantoic membrane xenograft model with the EGFR exon-19-deleted advanced NSCLC mutation. FEBS Open Bio 2025; 15:836-855. [PMID: 39887892 PMCID: PMC12051027 DOI: 10.1002/2211-5463.13970] [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: 08/28/2024] [Revised: 12/17/2024] [Accepted: 01/14/2025] [Indexed: 02/01/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) affects 10-50% of patients with epidermal growth factor receptor (EGFR) mutations. Osimertinib is a third-generation EGFR tyrosine kinase inhibitor (TKI) that radically changes the outcome of patients with tumors bearing EGFR sensitizing or EGFR T790M resistance mutations. However, resistance usually occurs, and new therapeutic combinations need to be explored. The chorioallantoic membrane (CAM) xenograft model is ideal for studying aggressive tumor growth and the responses to complex therapeutic combinations due to its vascularization and complex microenvironment. This study aims to demonstrate the relevance of analyzing a complex therapeutic response to osimertinib treatment, especially through advanced transcriptomic analysis with the CAM model, which has been limited thus far. We engrafted HCC827 cells (EGFR p.E746_A750del) into the CAM model and treated them with various osimertinib doses for 7 days. The study involved supervised multivariate discrimination and ontology analysis of human transcriptional data. We found that CDX tumor growth inversely correlated with osimertinib dosage, with a notable 35% tumor weight reduction at 10 μm. Transcriptomic analysis revealed that osimertinib reduces EGFR pathway activity and its effectors, and dampens chemotaxis, immune recruitment and angiogenesis, indicating that effectiveness extends beyond cellular mechanisms to the tissue level. This was supported by a 15% reduction in blood vessels around the xenograft in osimertinib-treated cases. This study is the first to demonstrate that ontological analysis of transcriptomic data in the CAM model aligns with clinical observations, highlighting the relevance of this methodology for understanding and ameliorating the efficacy of targeted therapy in NSCLC.
Collapse
Affiliation(s)
- David Barthélémy
- Department of Pharmacology‐Physiology‐Toxicology, Institute of Pharmaceutical and Biological Sciences of LyonUniversity Claude Bernard Lyon 1France
- Department of Biochemistry and Molecular BiologyLyon Sud Hospital, Hospices Civils de LyonPierre‐BéniteFrance
- Center for Innovation in Cancerology of Lyon (CICLY) Research Unit 3738, Faculty of Medicine and Maieutic Lyon SudUniversity Claude Bernard Lyon 1OullinsFrance
| | - Arnaud Vigneron
- INSERM U1052‐CNRS UMR5286, Comprehensive Cancer Center Léon Bérard, Cancer Research Center of Lyon, Institut Convergence PlascanUniversity Claude Bernard Lyon 1France
| | | | - Jérome Guitton
- Department of Pharmacology‐Physiology‐Toxicology, Institute of Pharmaceutical and Biological Sciences of LyonUniversity Claude Bernard Lyon 1France
- Department of Biochemistry and Molecular BiologyLyon Sud Hospital, Hospices Civils de LyonPierre‐BéniteFrance
| | - Emmanuel Grolleau
- Department of Acute Respiratory Disease and Thoracic OncologyLyon Sud Hospital, Hospices Civils de LyonPierre‐BéniteFrance
| | - Margaux Raffin
- Department of Biochemistry and Molecular BiologyLyon Sud Hospital, Hospices Civils de LyonPierre‐BéniteFrance
- Center for Innovation in Cancerology of Lyon (CICLY) Research Unit 3738, Faculty of Medicine and Maieutic Lyon SudUniversity Claude Bernard Lyon 1OullinsFrance
- Hospices Civils de Lyon, Circulating Cancer (CIRCAN) Program, Cancer InstitutePierre BéniteFrance
| | - Julie Balandier
- Department of Biochemistry and Molecular BiologyLyon Sud Hospital, Hospices Civils de LyonPierre‐BéniteFrance
- Center for Innovation in Cancerology of Lyon (CICLY) Research Unit 3738, Faculty of Medicine and Maieutic Lyon SudUniversity Claude Bernard Lyon 1OullinsFrance
- Hospices Civils de Lyon, Circulating Cancer (CIRCAN) Program, Cancer InstitutePierre BéniteFrance
| | - Gaëlle Lescuyer
- Department of Biochemistry and Molecular BiologyLyon Sud Hospital, Hospices Civils de LyonPierre‐BéniteFrance
- Center for Innovation in Cancerology of Lyon (CICLY) Research Unit 3738, Faculty of Medicine and Maieutic Lyon SudUniversity Claude Bernard Lyon 1OullinsFrance
- Hospices Civils de Lyon, Circulating Cancer (CIRCAN) Program, Cancer InstitutePierre BéniteFrance
| | - Mathilde Bardou
- Department of PathologyLyon Sud Hospital, Hospices Civils de LyonPierre‐BéniteFrance
| | - Florence Geiguer
- Department of Biochemistry and Molecular BiologyLyon Sud Hospital, Hospices Civils de LyonPierre‐BéniteFrance
- Center for Innovation in Cancerology of Lyon (CICLY) Research Unit 3738, Faculty of Medicine and Maieutic Lyon SudUniversity Claude Bernard Lyon 1OullinsFrance
- Hospices Civils de Lyon, Circulating Cancer (CIRCAN) Program, Cancer InstitutePierre BéniteFrance
| | - Sébastien Couraud
- Center for Innovation in Cancerology of Lyon (CICLY) Research Unit 3738, Faculty of Medicine and Maieutic Lyon SudUniversity Claude Bernard Lyon 1OullinsFrance
- Department of Acute Respiratory Disease and Thoracic OncologyLyon Sud Hospital, Hospices Civils de LyonPierre‐BéniteFrance
| | - Claire Bardel
- Department of BioinformaticsHospices Civils de LyonFrance
- Laboratory of Biometry and Evolutionary Biology, UMR 5558‐CNRSUniversity Claude Bernard Lyon 1VilleurbanneFrance
| | | | - Nazim Benzerdjeb
- Center for Innovation in Cancerology of Lyon (CICLY) Research Unit 3738, Faculty of Medicine and Maieutic Lyon SudUniversity Claude Bernard Lyon 1OullinsFrance
- Department of PathologyLyon Sud Hospital, Hospices Civils de LyonPierre‐BéniteFrance
| | - Léa Payen
- Department of Pharmacology‐Physiology‐Toxicology, Institute of Pharmaceutical and Biological Sciences of LyonUniversity Claude Bernard Lyon 1France
- Department of Biochemistry and Molecular BiologyLyon Sud Hospital, Hospices Civils de LyonPierre‐BéniteFrance
- Center for Innovation in Cancerology of Lyon (CICLY) Research Unit 3738, Faculty of Medicine and Maieutic Lyon SudUniversity Claude Bernard Lyon 1OullinsFrance
- Hospices Civils de Lyon, Circulating Cancer (CIRCAN) Program, Cancer InstitutePierre BéniteFrance
| |
Collapse
|
26
|
Ye YT, Xia HY, Li J, Wang SB, Chen AZ, Kankala RK. Nanoarchitecting intelligently encapsulated designs for improved cancer therapy. Front Bioeng Biotechnol 2025; 13:1587178. [PMID: 40375976 PMCID: PMC12078215 DOI: 10.3389/fbioe.2025.1587178] [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: 03/04/2025] [Accepted: 04/14/2025] [Indexed: 05/18/2025] Open
Abstract
Despite the success in exploring various aspects of origination and therapeutic strategies, cancer has remained one of the most dreadful metabolic disorders due to failure to eradicate tumors comprehensively and frequent recurrence because of acquired resistance to the drugs. Recently, several advancements have been evidenced in the fabrication of various smart nanocarriers encapsulated with multiple components. Several reasons for smart nanoencapsulation include the enhancement of the bioavailability of drugs, precise targetability to reduce adverse effects on normal cells, and the ability to enable controlled drug release rates at the tumor sites. In addition, these smart nanocarriers protect encapsulated therapeutic cargo from deactivation, responsively delivering it based on the physiological or pathological characteristics of tumors. In this review, we present various smart approaches for cancer therapy, including organic materials, inorganic components, and their composites, as well as biomembrane-based nanoencapsulation strategies. These nanoencapsulation strategies, along with practical applications and their potential in cancer treatment, are discussed in depth, highlighting advantages and disadvantages, as well as aiming to reveal the ultimate prospects of nanoencapsulation in enhancing drug delivery efficiency and targeted cancer therapy.
Collapse
Affiliation(s)
- Ying-Tong Ye
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, China
| | - Hong-Ying Xia
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, China
| | - Jie Li
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, China
- College of Chemical Engineering, Huaqiao University, Xiamen, China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, China
- College of Chemical Engineering, Huaqiao University, Xiamen, China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, China
| |
Collapse
|
27
|
Abu-Serie MM, Gutiérrez-García AK, Enman M, Vaish U, Fatima H, Dudeja V. Ferroptosis- and stemness inhibition-mediated therapeutic potency of ferrous oxide nanoparticles-diethyldithiocarbamate using a co-spheroid 3D model of pancreatic cancer. J Gastroenterol 2025; 60:641-657. [PMID: 39888413 PMCID: PMC12014774 DOI: 10.1007/s00535-025-02213-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/10/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with a high mortality rate and exhibits a limited response to apoptosis-dependent chemotherapeutic drugs (e.g., gemcitabine, Gem). This is mainly attributed to the antioxidant defense system (glutathione and aldehyde dehydrogenase (ALDH) 1A1), which sustains stemness features of cancer stem cells (CSCs) and activated pancreatic stellate cells (PSCs)-generated excess stromal proteins. This dense stroma retards drug delivery. METHODS This study established co-spheroid model consisting of mouse PDAC cell line (KPC) and PSCs (1:5) to accurately investigate the anti-PDAC activity of nanocomplex of ferrous oxide nanoparticles-diethyldithiocarbamate (FeO NPs-DE), compared to Gem, using in vitro and in vivo 3D models. RESULTS In vitro and in vivo co-spheroid models demonstrated higher therapeutic efficacy of FeO NPs-DE than Gem. FeO NPs-DE induced selective accumulation of iron-dependent ferroptosis (non-apoptosis)-generated a lethal lipid peroxidation that was potentiated by DE-mediated glutathione and ALDH1A1 suppression. This led to collapse of stemness, as evidenced by down-regulating CSC genes and p-AKT protein expression. Subsequently, gene and/or protein levels of PSC activators (transforming growth factor (TGF)-β, plasminogen activator inhibitor-1, ZEB1, and phosphorylated extracellular signal-regulated kinase) and stromal proteins (collagen 1A2, smooth muscle actin, fibronectin, and matrix metalloproteinase-9) were suppressed. Moreover, DE of nanocomplex enhanced caspase 3-dependent apoptosis with diminishing the main oncogene, BCL-2. CONCLUSIONS FeO NPs-DE had a stronger eradicating effect than Gem on primary and metastatic peritoneal PDAC tumors. This nanocomplex-mediated ferroptosis and stemness inhibition provides an effective therapeutic approach for PDAC.
Collapse
Affiliation(s)
- Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El‑Arab City, Alexandria, 21934, Egypt.
| | - Ana K Gutiérrez-García
- Division of Surgical Oncology, Department of Surgery, University of Alabama at Birmingham (UAB), Birmingham, Alabama, 35294, USA
| | - Macie Enman
- Division of Surgical Oncology, Department of Surgery, University of Alabama at Birmingham (UAB), Birmingham, Alabama, 35294, USA
| | - Utpreksha Vaish
- Division of Surgical Oncology, Department of Surgery, University of Alabama at Birmingham (UAB), Birmingham, Alabama, 35294, USA
| | - Huma Fatima
- Department of Pathology, Division of Anatomic Pathology, University of Alabama at Birmingham (UAB), Birmingham, Alabama, 35249, USA
| | - Vikas Dudeja
- Division of Surgical Oncology, Department of Surgery, University of Alabama at Birmingham (UAB), Birmingham, Alabama, 35294, USA
| |
Collapse
|
28
|
Feng P, Zhang X, Gao J, Jiang L, Li Y. The Roles of Exosomes in Anti-Cancer Drugs. Cancer Med 2025; 14:e70897. [PMID: 40298189 PMCID: PMC12038748 DOI: 10.1002/cam4.70897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Cancer is an escalating global health issue, with rising incidence rates annually. Chemotherapy, a primary cancer treatment, often exhibits low tumor-targeting efficiency and severe side effects, limiting its effectiveness. Recent research indicates that exosomes, due to their immunogenicity and molecular delivery capabilities, hold significant potential as drug carriers for tumor treatment. METHODS This review summarizes the current status, powerful therapeutic potential, and challenges of using exosomes for the treatment of tumors. RESULTS Exosomes are crucial in tumor diagnosis, onset, and progression. To improve the efficacy of exosome-based treatments, researchers are exploring various biological, physical, and chemical approaches to engineer exosomes as a new nanomedicine translational therapy platform with broad and alterable therapeutic capabilities. Numerous clinical trials are currently underway investigating the safety and tolerability of exosomes carrying drugs to specific sites for the treatment of tumors. CONCLUSIONS Exosomes can be engineered as carriers to deliver therapeutic molecules to specific cells and tissues, offering a novel approach for disease treatment.
Collapse
Affiliation(s)
- Panpan Feng
- Department of RadiotherapyThe First Affiliated Hospital of Jinzhou Medical UniversityJinzhouChina
| | - Xiaodong Zhang
- Department of General SurgeryBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Jian Gao
- Science Experiment Center of China Medical UniversityShenyangChina
| | - Lei Jiang
- Department of General SurgeryThe First Affiliated Hospital of Jinzhou Medical UniversityJinzhouChina
| | - Yan Li
- Department of RadiotherapyThe First Affiliated Hospital of Jinzhou Medical UniversityJinzhouChina
- Liaoning Provincial Key Laboratory of Clinical Oncology MetabonomicsThe First Affiliated Hospital of Jinzhou Medical UniversityJinzhouChina
| |
Collapse
|
29
|
Jafary F, Moradi A, Ganjalikhany MR, Hassanpour Dehnavi A, Seifati SM, Khodarahmi A, Hemati M. Investigation of the inhibitory peptide effect as novel strategy in cancer treatment: Targeting the tetramerization of lactate dehydrogenase A. Int J Biol Macromol 2025; 306:140878. [PMID: 39988151 DOI: 10.1016/j.ijbiomac.2025.140878] [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: 08/07/2024] [Revised: 02/02/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
The Warburg effect is detected in numerous types of solid tumors and is characterized by the enhancement of glucose consumption and an increase in the rate of glycolysis in cancer cells. Lactate dehydrogenase A (LDHA) plays a critical role in the Warburg effect and to date, a number of LDHA inhibitors have been identified that act as competitive inhibitors of both the substrate pyruvate and the coenzyme NADH. In our previous study, a set of novel peptides were designed using in silico structure-based method with the aim of inhibiting the activity of LDHA by disrupting the interaction between its enzyme subunits. In the present study employed in vitro and in vivo techniques, to explore the influence of these peptides on the induction of cytotoxicity effect on cancer cell lines. Our findings revealed that the designed peptide effectively inhibits LDHA, leading to the induction of apoptosis and G2/M cell cycle arrest. Finally, we also analyzed the impact of the designed peptide on Balb/c mice, confirming the results of in silico and in vitro studies. Ultimately, the designed peptide had the ability to induce apoptosis in cancer cells by LDHA inhibition.
Collapse
Affiliation(s)
- Farzaneh Jafary
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran; Clinical Research Development Center, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Ali Moradi
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran.
| | - Mohamad Reza Ganjalikhany
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Azam Hassanpour Dehnavi
- Department of Anatomical Sciences, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran; Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mohammad Seifati
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ameneh Khodarahmi
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mahdie Hemati
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran; Medical Nanotechnology and Tissue Engineering Research Center, Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
30
|
Xie T, Zhang Y, Ling N, Yuan Y, Liu W, Guo J, Wei X, Peng T, Wang Z, Dai J, Chen W, Yan M, Wu X, Wu W, Ye M, Tan W. Aptamer as a Molecular Tethering Agent Induces PrP C Aggregation and Degradation to Inhibit Melanoma Proliferation. Angew Chem Int Ed Engl 2025:e202425051. [PMID: 40304120 DOI: 10.1002/anie.202425051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/05/2025] [Accepted: 04/29/2025] [Indexed: 05/02/2025]
Abstract
Melanoma, a malignant tumor originating from melanocytes, is the most aggressive and deadly form of skin cancer. Previous studies have revealed that the cellular prion protein (PrPC) is frequently overexpressed in melanoma, contributing to tumor progression. This study presents the first proof of concept evidence that nucleic acid aptamers can be used to construct a molecular tethering agent that regulates PrPC protein levels by inducing membrane-bound PrPC aggregation for antimelanoma therapy. Using a screening strategy combining cell-SELEX and cell-internalization SELEX, we obtained ssDNA aptamer, TT-1e, specifically binding to melanoma cells and tissues. We identified that the binding site of TT-1e is located at the octapeptide repeat region of glycosylated PrPC. Based on the binding characteristics of TT-1e, we engineered an aptamer-based molecular tethering agent TTe-TTe. We found that TTe-TTe induces aggregation of cell surface PrPC, promoting its internalization and facilitating its lysosomal degradation. This process resulted in the inhibition of AKT pathway activation. Importantly, in vivo studies confirmed the ability of TTe-TTe to target melanoma xenografts and suppress tumor growth through this unique mechanism. Our study presents a promising strategy for targeted melanoma therapy and introduces a paradigm-shifting approach for manipulating protein levels using aptamers as molecular tethering agents.
Collapse
Affiliation(s)
- Tiantian Xie
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Yibin Zhang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Neng Ling
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China, State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China, Zhejiang Key Laboratory of Key Technologies for Visual Pathway Reconstruction,Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yijun Yuan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Weibin Liu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China, State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China, Zhejiang Key Laboratory of Key Technologies for Visual Pathway Reconstruction,Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Junxiao Guo
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Xianhua Wei
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Tianhuan Peng
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Zhiyan Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Jing Dai
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Wenjing Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Ming Yan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Xiaoqiu Wu
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Wencan Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China, State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China, Zhejiang Key Laboratory of Key Technologies for Visual Pathway Reconstruction,Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology,College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
31
|
Connelly KE, Hullin K, Abdolalizadeh E, Zhong J, Eiser D, O'Brien A, Collins I, Das S, Duncan G, Chanock SJ, Stolzenberg-Solomon RZ, Klein AP, Wolpin BM, Hoskins JW, Andresson T, Smith JP, Amundadottir LT. Allelic effects on KLHL17 expression underlie a pancreatic cancer genome-wide association signal at chr1p36.33. Nat Commun 2025; 16:4055. [PMID: 40307206 PMCID: PMC12044007 DOI: 10.1038/s41467-025-59109-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is the third leading cause of cancer-related deaths in the U.S. Both rare and common germline variants contribute to PDAC risk. Here, we fine-map and functionally characterize a common PDAC risk signal at chr1p36.33 (tagged by rs13303010) identified through a genome wide association study (GWAS). One of the fine-mapped SNPs, rs13303160 (OR = 1.23 (95% CI 1.15-1.32), P-value = 2.74×10-9, LD r2 = 0.93 with rs13303010 in 1000 G EUR samples) demonstrated allele-preferential gene regulatory activity in vitro and binding of JunB and JunD in vitro and in vivo. Expression Quantitative Trait Locus (eQTL) analysis identified KLHL17 as a likely target gene underlying the signal. Proteomic analysis identified KLHL17 as a member of the Cullin-E3 ubiquitin ligase complex with vimentin and nestin as candidate substrates for degradation in PDAC-derived cells. In silico differential gene expression analysis of high and low KLHL17 expressing GTEx pancreas samples suggested an association between lower KLHL17 levels (risk associated) and pro-inflammatory pathways. We hypothesize that KLHL17 may mitigate cell injury and inflammation by recruiting nestin and vimentin for ubiquitination and degradation thereby influencing PDAC risk.
Collapse
Affiliation(s)
- Katelyn E Connelly
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
| | - Katherine Hullin
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Ehssan Abdolalizadeh
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Jun Zhong
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Daina Eiser
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Aidan O'Brien
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Irene Collins
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Sudipto Das
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Frederick, MD, USA
| | - Gerard Duncan
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Frederick, MD, USA
| | - Stephen J Chanock
- Laboratory of Genomic Susceptibility, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Rachael Z Stolzenberg-Solomon
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Alison P Klein
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jason W Hoskins
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Thorkell Andresson
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Frederick, MD, USA
| | - Jill P Smith
- Department of Medicine, Georgetown University, Washington, USA
| | - Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
| |
Collapse
|
32
|
Wang L, Zhao X, Zhu W, Ji Y, Zeng M, Wang M. Development and validation of a CT-based nomogram to preoperative prediction of pancreatic neuroendocrine tumors (pNETs) grade. Abdom Radiol (NY) 2025:10.1007/s00261-025-04959-z. [PMID: 40293523 DOI: 10.1007/s00261-025-04959-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND/PURPOSE It is challenging to determine the pancreatic neuroendocrine tumors (pNETs) malignancy grade noninvasively. We aim to establish a CT - based diagnostic nomogram to predict the tumor grade of pNETs. METHODS The patients with pathologically confirmed pNETs were recruited in two centers between January 2009 and November 2020. PNETs were subdivided into three grades according to the 2017 World Health Organization classification: low-grade G1 NETs, intermediate-grade G2 NETs, and high-grade G3 NETs. The features on the CT images were carefully evaluated. To build the nomogram, multivariable logistic regression analysis was performed on the imaging features selected by LASSO to generate a combined indicator for estimating the tumor grade. RESULTS A total of 162 pNETs (training set n = 114, internal validation set n = 21, external validation set, n = 48) were admitted, including 73 (45.1%) G1 and 89 (54.9%) G2/3. A nomogram comprising the tumor margin, tumor size, neuroendocrine symptoms and the enhanced ratio on portal vein phase images of tumor was established to predict the malignancy grade of pNETs. The mean AUC for the nomogram was 0.848 (95% CI, 0.918-0.953). Application of the developed nomogram in the internal validation dataset still yielded good discrimination (AUC, 0.835; 95% CI, 0.915-0.954). The externally validated nomogram yielded a slightly lower AUC of 0.770 (95% CI, 0.776-0.789). CONCLUSIONS The nomogram model demonstrated good performance in preoperatively predicting the malignancy grade of pNETs, and can provide clinicians with a simple, practical, and non-invasive tool for personalized management of pNETs patients.
Collapse
Affiliation(s)
- Liangqi Wang
- Department of Radiology, Shanghai Geriatric Medical Center, Shanghai, China
| | - Xiangtian Zhao
- Department of Radiology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Wenxia Zhu
- Department of Radiology, The Third People's Hospital of Qingdao, Qingdao, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mingliang Wang
- Department of Radiology, Shanghai Geriatric Medical Center, Shanghai, China.
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
33
|
Zhi Y, Guo Y, Li S, He X, Wei H, Laster K, Wu Q, Zhao D, Xie J, Ruan S, Lemoine NR, Li H, Dong Z, Liu K. FBL promotes hepatocellular carcinoma tumorigenesis and progression by recruiting YY1 to enhance CAD gene expression. Cell Death Dis 2025; 16:348. [PMID: 40289107 PMCID: PMC12034760 DOI: 10.1038/s41419-025-07684-z] [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: 12/04/2024] [Revised: 04/13/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Accumulating evidence suggests that epigenetic dysregulation contributes to the initiation and progression of HCC. We aimed to investigate key epigenetic regulators that contribute to tumorigenesis and progression, providing a theoretical basis for targeted therapy for HCC. We performed a comprehensive epigenetic analysis of differentially expressed genes in LIHC from the TCGA database. We identified fibrillarin (FBL), an rRNA 2'-O-methyltransferase, as an essential contributor to HCC. A series of in vitro and in vivo biological experiments were performed to investigate the potential mechanisms of FBL. FBL knockdown suppressed the proliferation of HCC cells. In vivo studies using cell-derived xenograft (CDX), patient-derived xenograft (PDX), and diethylnitrosamine (DEN)-induced HCC models in Fbl liver-specific knockout mice demonstrated the critical role of FBL in HCC carcinogenesis and progression. Mechanistically, FBL regulates the expression of CAD in HCC cells by recruiting YY1 to the CAD promoter region. We also revealed that fludarabine phosphate is a novel inhibitor of FBL and can inhibit HCC growth in vitro and in vivo. The antitumor activity of lenvatinib has been shown to be synergistically enhanced by fludarabine phosphate. Our study highlights the cancer-promoting role of the FBL-YY1-CAD axis in HCC and identifies fludarabine phosphate as a novel inhibitor of FBL. A schematic diagram depicting the FBL-YY1-CAD signaling pathway and its regulatory role in HCC progression.
Collapse
Affiliation(s)
- Yafei Zhi
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, China
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, China
- Cancer Chemistry International Collaboration Laboratory, Zhengzhou, China
| | - Yan Guo
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Shiliang Li
- Innovation Center for AI and Drug Discovery, East China Normal University, Shanghai, China
| | - Xinyu He
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Huifang Wei
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Kyle Laster
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Qiong Wu
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Dengyun Zhao
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Jinxin Xie
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Shanshan Ruan
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Nicholas R Lemoine
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy; The School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China.
- Center for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK.
| | - Honglin Li
- Innovation Center for AI and Drug Discovery, East China Normal University, Shanghai, China.
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China.
- Lingang Laboratory, Shanghai, China.
| | - Zigang Dong
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, China.
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, China.
- Cancer Chemistry International Collaboration Laboratory, Zhengzhou, China.
| | - Kangdong Liu
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, China.
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, China.
- Cancer Chemistry International Collaboration Laboratory, Zhengzhou, China.
| |
Collapse
|
34
|
Xu M, Zhang QT, Zhou L, Cai YW, Liu H, Zhao QL, Tian JH, Huang YK, Ren P, Huang X. Ferulic acid in Chaihu Shugan San modulates depression-like behavior, endothelial and gastrointestinal dysfunction in rats via the Ghrl-Edn1/Mecp2/P-mTOR/VEGFA pathway: A multi-omics study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119624. [PMID: 40127829 DOI: 10.1016/j.jep.2025.119624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/16/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In a global context of escalating multimorbidity, characterized by the co-occurrence of major depressive disorder (MDD), endothelial dysfunction (ED), and gastrointestinal dysregulation (GD), the quest for effective treatments has become paramount. Central to these interconnected conditions is oxidative stress (OS), a pivotal factor that has been extensively studied yet remains inadequately addressed. This study introduces Chaihu-Shugan-San (CSS) and its absorbed component ferulic acid (FA), a potent antioxidant derived from medicinal plants, as a novel therapeutic approach with the unique ability to counter the multifaceted effects of acute forced swimming (AFS)-induced depression, ED, and GD. Unlike traditional single-disease-focused studies, our research explores the synergistic effects of CSS and FA across these interrelated disorders, offering a groundbreaking perspective. AIM OF THE STUDY This study aims to evaluate CSS and FA in treating depression-related multimorbidity triggered by AFS and to uncover the shared underlying mechanisms of FA. MATERIALS AND METHODS A depression-like model in rats was induced by AFS, and an OS model was established in endothelial cells (ECs) through hydrogen peroxide treatment. We investigated the effects of CSS and FA on MDD, ED, and GD in rats and OS levels in ECs. Our assessments included hematoxylin and eosin (HE) staining, biochemical assays, and behavioral studies. We conducted an integrated analysis of transcriptomics, proteomics, and phosphoproteomics data to elucidate the underlying mechanisms. The identification of relevant targets was confirmed through Western blotting (WB), real-time quantitative polymerase chain reaction (RT-qPCR), molecular docking studies, and an extensive literature review. RESULTS Our findings indicate that CSS and FA not only significantly mitigate AFS-induced abnormalities in the open field test (OFT), forced swim test (FST), and related behaviors such as gastric emptying and intestinal transit in rats but also ameliorate depression, ED, GD, inflammation and OS-related biomarker levels, alongside HE staining in gastric sinus and aorta slices. The study also highlights that FA can influence OS and endothelial function in ECs. Moreover, a combined multi-omics analysis unveiled several OS-related pathways, including the mTOR and p53 signaling pathways. Our research elucidates that the Ghrl-Edn1/Mecp2/P-mTOR/Vegfa-associated OS signaling pathway is pivotal in countering AFS-induced multimorbidity, expanding beyond the conventional disease-specific focus. CONCLUSIONS This pioneering study underscores capability of CSS and FA to tackle AFS-induced multimorbidity concurrently and intricately details FA's antioxidative mechanisms within ECs. The insights gleaned offer a novel perspective on FA's role in multimorbidity regulation and its potential to modulate OS, especially in the complex environment of ECs. Given the urgent global health challenges, our research positions FA as a promising therapeutic contender, advocating for a paradigm shift in multimorbidity management.
Collapse
Affiliation(s)
- Min Xu
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qian-Tao Zhang
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Zhou
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ya-Wen Cai
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hao Liu
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiu-Long Zhao
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing-Hua Tian
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun-Ke Huang
- Women's Hospital, Zhejiang University School of Medicine, Gynecology Department, Zhejiang, China
| | - Ping Ren
- Department of Geriatrics, Jiangsu Province Hospital of TCM, Nanjing, China
| | - Xi Huang
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
35
|
Núñez JC, Rivera MT, Stevens MA. Adenocarcinoma of the duodenal papilla with synchronous peritoneal metastases-5 years of overall survival: A case report. World J Clin Oncol 2025; 16:103651. [PMID: 40290683 PMCID: PMC12019277 DOI: 10.5306/wjco.v16.i4.103651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/20/2024] [Accepted: 02/08/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Ampullary adenocarcinomas are a rare disease. They can be classified anatomically or according to their histology into intestinal, pancreatobiliary, and mixed subtypes, with different subtypes having distinct prognoses and potential treatments. We report a clinical case of a patient with mixed type adenocarcinoma of the ampulla of Vater, with predominantly intestinal histology, associated with an isolated and synchronous peritoneal carcinomatosis. It is the only case reported in the literature of duodenal ampulla cancer with synchronous peritoneal metastases, with long-term survival. CASE SUMMARY A 53-year-old male patient with non-insulin-dependent diabetes presented with acute abdominal pain in the right hypochondrium. Images revealed dilatation of the biliary tract and the duct of Wirsung, without a clear obstructive factor. Upper gastrointestinal endoscopy revealed a tumor in the duodenal papilla. Biopsies confirmed an adenocarcinoma. In the first surgical step, a biliodigestive bypass was performed in association with resection of the carcinomatosis. Peritoneal metastases was found during the intraoperative period. Subsequently, chemotherapy with the folinic acid, fluorouracil, and oxaliplatin regimen was administered based on histology, and a favorable response was achieved. After a multidisciplinary discussion, the Whipple procedure was performed. A delayed biopsy showed disease-free margins. The patient achieved 5 years of overall survival in August 2024, and 4 years of disease-free survival in September 2024. CONCLUSION We conclude that an important value of this work is showing individualized treatment for a patient with cancer.
Collapse
Affiliation(s)
- Julio César Núñez
- Department of Surgery, Hospital del Salvador, Santiago 7500922, Región Metropolitana, Chile
- Department of Surgery, Clínica Dávila-Vespucio, Santiago 8241479, Región Metropolitana, Chile
| | - María Teresa Rivera
- Department of Anatomic Pathology, Hospital del Salvador, Santiago 7500922, Región Metropolitana, Chile
| | - Mary Ann Stevens
- Department of Medical Oncology, Hospital del Salvador, Santiago 7500922, Región Metropolitana, Chile
- Department of Medical Oncology, Clínica Alemana, Santiago 7650568, Región Metropolitana, Chile
| |
Collapse
|
36
|
Michetti F, Cirone M, Strippoli R, D'Orazi G, Cordani M. Mechanistic insights and therapeutic strategies for targeting autophagy in pancreatic ductal adenocarcinoma. Discov Oncol 2025; 16:592. [PMID: 40266451 PMCID: PMC12018664 DOI: 10.1007/s12672-025-02400-x] [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: 12/24/2024] [Accepted: 04/15/2025] [Indexed: 04/24/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterised by early metastasis and resistance to anti-cancer therapy, leading to an overall poor prognosis. Macroautophagy (hereinafter referred to as autophagy) is a conserved cellular homeostasis mechanism that degrades various cargoes (e.g., proteins, organelles, and pathogens) mainly playing a role in promoting survival under environmental stress. Autophagy is an essential defense mechanism against PDAC initiation, acting on multiple levels to maintain cellular and tissue homeostasis. However, autophagy is also intimately involved in the molecular mechanisms driving PDAC progression, facilitating the adaptation of cancer cells to the tumor microenvironment's harsh conditions. In this review, we examine the complex role of autophagy in PDAC and assess the potential of modulating autophagy as a therapeutic strategy. By reviewing current research and clinical trials, we seek to elucidate how targeting autophagy can disrupt PDAC tumor survival mechanisms, enhance the efficacy of existing treatments, and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Federica Michetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149, Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149, Rome, Italy.
| | - Gabriella D'Orazi
- UniCamillus-Saint Camillus International University of Health and Medical Sciences, Via di Sant'Alessandro 8, 00131, Rome, Italy.
- Department of Research and Advanced Technologies, Regina Elena National Cancer Institute IRCCS, Via Elio Chianesi 51, 00144, Rome, Italy.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040, Madrid, Spain.
| |
Collapse
|
37
|
Bai S, Hu Y, Chen N, Zhou L, Ju W, Qiao X, Yu J. DCAF13 influences breast cancer chemotherapy resistance through metabolic reprogramming by regulating c-Myc expression. Med Oncol 2025; 42:178. [PMID: 40268788 DOI: 10.1007/s12032-025-02722-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Chemotherapy resistance remains a major obstacle in breast cancer treatment. This study identifies DCAF13, a substrate recognition receptor of the CRL4 ubiquitin ligase complex, as a critical regulator of chemotherapy resistance through c-Myc-driven metabolic reprogramming. We found that DCAF13 expression was significantly upregulated in chemotherapy-resistant breast cancer cell lines compared to their parental counterparts. Inhibition of DCAF13 enhanced chemotherapy sensitivity, whereas its overexpression suppressed drug sensitivity. Mechanistically, DCAF13 upregulated c-Myc expression, driving metabolic reprogramming, characterized by increased glycolysis and oxidative phosphorylation. This metabolic shift promoted cell proliferation and resistance to chemotherapy. Clinically, DCAF13 expression correlated with poor prognosis in breast cancer patients, particularly in advanced stages and triple-negative breast cancer (TNBC). Our findings highlight the DCAF13-c-Myc axis as a critical mediator of chemotherapy resistance, suggesting that targeting this pathway could provide novel therapeutic strategies to overcome drug resistance in breast cancer. Further clinical studies are needed to explore the potential of DCAF13 as a therapeutic target.
Collapse
Affiliation(s)
- Shiqiang Bai
- Department of Pathology, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, 118000, China
| | - Yunlong Hu
- Department of Emergency and Intensive Care Unit, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, 118000, China
| | - Ning Chen
- Department of Pathology, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, 118000, China
| | - Liang Zhou
- Department of General Surgery, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, 118000, China
| | - Weiwei Ju
- Medical College of Liaodong University, Dandong, 118003, China
| | - Xinyi Qiao
- Department of General Surgery, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, 118000, China
| | - Jianyu Yu
- Department of General Surgery, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, 118000, China.
| |
Collapse
|
38
|
Awale S, Maneenet J, Phan ND, Nguyen HH, Fujii T, Ihmels H, Soost D, Tajuddeen N, Feineis D, Bringmann G. Toyaburgine, a Synthetic N-Biphenyl-Dihydroisoquinoline Inspired by Related N, C-Coupled Naphthylisoquinoline Alkaloids, with High In Vivo Efficacy in Preclinical Pancreatic Cancer Models. ACS Chem Biol 2025; 20:917-929. [PMID: 40048237 DOI: 10.1021/acschembio.4c00870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Pancreatic cancer is a highly aggressive and lethal malignancy, with a 5-year survival rate below 10%. Traditional chemotherapy, including gemcitabine, has limited efficacy due to chemoresistance and a unique tumor microenvironment characterized by hypovascularity and nutrient deprivation. This study reports on the discovery of a new N-biphenyl-dihydroisoquinoline, named toyaburgine (4), inspired by naturally occurring N,C-coupled naphthylisoquinoline alkaloids. Developed through systematic structural optimization, toyaburgine is a potent anticancer agent, showing promise for pancreatic cancer treatment. It exhibits strong antiausterity activity with low nanomolar PC50 values, effectively inhibiting pancreatic cancer cell viability under nutrient-deprived conditions. In vitro, 4 causes significant morphological changes and cancer cell death in MIA PaCa-2 cells while also inhibiting cell migration and colony formation, which indicates its antimetastatic potential. Mechanistically, toyaburgine disrupts the PI3K/Akt/mTOR pathway, essential for pancreatic cancer cell survival in a stressful microenvironment, and inhibits MIA PaCa-2 spheroid formation. In vivo, toyaburgine, alone or combined with gemcitabine, shows effective tumor suppression in subcutaneous xenograft and clinically relevant orthotopic models, where it also reduces cachexia. These results highlight the potential of toyaburgine as a new therapeutic drug for pancreatic cancer. Its combination with gemcitabine presents a promising treatment approach by targeting both proliferating and gemcitabine-resistant cancer cells.
Collapse
Affiliation(s)
- Suresh Awale
- Natural Drug Discovery Laboratory, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Juthamart Maneenet
- Natural Drug Discovery Laboratory, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Nguyen Duy Phan
- Natural Drug Discovery Laboratory, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Hung Hong Nguyen
- Natural Drug Discovery Laboratory, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Tsutomu Fujii
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Heiko Ihmels
- Department of Chemistry-Biology, and Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), University of Siegen, Adolf-Reichwein-Straße 2, 57068 Siegen, Germany
| | - Denisa Soost
- Department of Chemistry-Biology, and Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), University of Siegen, Adolf-Reichwein-Straße 2, 57068 Siegen, Germany
| | - Nasir Tajuddeen
- Department of Chemistry-Biology, and Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), University of Siegen, Adolf-Reichwein-Straße 2, 57068 Siegen, Germany
- Department of Chemistry, Ahmadu Bello University, 810107 Zaria, Nigeria
| | - Doris Feineis
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Gerhard Bringmann
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
39
|
Indelicato S, Bongiorno D, Mauro M, Cascioferro S. Recent Developments of 1,3,4-Thiadiazole Compounds as Anticancer Agents. Pharmaceuticals (Basel) 2025; 18:580. [PMID: 40284015 PMCID: PMC12030488 DOI: 10.3390/ph18040580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
The World Health Organization has recently underlined the increasing global burden of cancer, with a particularly alarming impact on underserved populations. In recent years, 1,3,4-thiadiazole has emerged as a versatile pharmacophore to obtain bioactive compounds. The pharmacological properties of this ring are primarily attributed to its role as a bioisostere of pyrimidine, the core structure of three nucleic bases. This structural feature endows 1,3,4-thiadiazole derivatives with the ability to interfere with DNA replication processes. Additionally, the mesoionic behavior of this heterocycle gives it important properties, such as the ability to cross biological membranes and interact with target proteins. Noteworthy, in analogy to the other sulfur heterocycles, the presence of C-S σ* orbitals, conferring small regions of low electron density on the sulfur atom, makes interaction with the target easier. This review focuses on the most promising anticancer agents with 1,3,4-thiadiazole structure reported in the past five years, providing information that may be useful to medicinal chemists who intend to develop new anticancer derivatives.
Collapse
Affiliation(s)
| | | | | | - Stella Cascioferro
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy; (S.I.); (D.B.); (M.M.)
| |
Collapse
|
40
|
Sui Y, Shen Z, Pan R, Ma R, Si R, Feng J, Zhou G. AHSA1-HSP90AA1 complex stabilized IFI6 and TGFB1 promotes mitochondrial stability and EMT in EGFR-mutated lung adenocarcinoma under Osimertinib pressure. Cell Death Dis 2025; 16:298. [PMID: 40234395 PMCID: PMC12000569 DOI: 10.1038/s41419-025-07650-9] [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: 08/12/2024] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
Tyrosine kinase inhibitors (TKIs) have substantially improved the management of lung adenocarcinoma harboring epidermal growth factor receptor (EGFR) mutations, however, not all patients can derive benefit from it. We found that the overexpression of IFI6 under the influence of the AHSA1-HSP90AA1 complex significantly enhances Osimertinib resistance in EGFR-mutated lung adenocarcinoma cells. This effect is achieved by stabilizing mitochondrial function, reducing apoptosis, and promoting cell survival pathways via increased Akt phosphorylation. Additionally, we revealed that TGFB1 further promotes epithelial-mesenchymal transition (EMT) and enhances the invasive and migratory capabilities of these cells, thereby intensifying resistance. Regarding mechanisms, the AHSA1-HSP90AA1 complex stabilizes IFI6 and TGFB1 to enhance cell survival and Osimertinib resistance in EGFR mutant lung adenocarcinoma. IFI6 not only aids in cellular survival under drug stress but also promotes aggressive tumor phenotypes, suggesting its viability as a novel biomarker and therapeutic target for overcoming primary TKI resistance.
Collapse
Affiliation(s)
- Ying Sui
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Ziyang Shen
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Rongtian Pan
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Rong Ma
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Rujia Si
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Jifeng Feng
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China.
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China.
| | - Guoren Zhou
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China.
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China.
| |
Collapse
|
41
|
Luo W, Xu M, Wong N, Ng CSH. Alternative Splicing in Lung Adenocarcinoma: From Bench to Bedside. Cancers (Basel) 2025; 17:1329. [PMID: 40282505 PMCID: PMC12025742 DOI: 10.3390/cancers17081329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is a highly heterogeneous tumor and the most prevalent pathological type of lung cancer. The alternative splicing (AS) of mRNA enables the generation of multiple protein products from a single gene. This is a tightly regulated process that significantly contributes to the proteome diversity in eukaryotes. Recent multi-omics studies have delineated the splicing profiles that underline LUAD tumorigenesis from initiation to metastasis. Such progress holds robust promise to facilitate the development of screening strategies and individualized therapies. Perturbed AS fosters the emergence of novel neoantigen resources and disturbances in the immune microenvironment, which allow new investigations into modulatory targets for LUAD immunotherapy. This review presents an update on the landscape of dysregulated splicing events in LUAD and the associated mechanisms and theranostic perspectives with unique insights into AS-based immunotherapy, such as Chimeric Antigen Receptor T cell therapy. These AS variants can be used in conjunction with current therapeutic modules in LUAD, allowing bench to bedside translation to combat this highly malignant cancer.
Collapse
Affiliation(s)
| | | | - Nathalie Wong
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China; (W.L.); (M.X.)
| | - Calvin Sze-Hang Ng
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China; (W.L.); (M.X.)
| |
Collapse
|
42
|
Yi AQ, Xie GH. Pancreatic neuroendocrine neoplasms coexisting with biliary intraductal papillary mucinous neoplasm: A case report and review of literature. World J Gastrointest Oncol 2025; 17:100497. [PMID: 40235901 PMCID: PMC11995319 DOI: 10.4251/wjgo.v17.i4.100497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/06/2024] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Pancreatic neuroendocrine neoplasms (pNENs) are rare, heterogeneous tumors accounting for 1%-2% of pancreatic tumors, with significant malignant potential. Intraductal papillary mucinous neoplasm of the bile duct (IPMN-B) is a rare precancerous lesion in the bile duct system, with potential for malignancy. The combination of pNENs and IPMN-B is exceptionally rare and often leads to misdiagnosis. This study aims to report a rare case of pNENs combined with IPMN-B treated at Yanbian University Hospital to improve understanding and management of this unusual tumor combination. CASE SUMMARY We retrospectively analyzed a case from Yanbian University Hospital. We reviewed clinical records, imaging findings, endoscopic retrograde cholangiopancreatography, surgical exploration, and histopathological examination. The patient was diagnosed with pNENs and IPMN-B. Surgical treatment was performed, with follow-up showing effective management and no significant recurrence. CONCLUSION This case represents the first report of pNENs combined with IPMN-B. It highlights the need for thorough diagnostic evaluation to prevent misdiagnosis and improve treatment strategies.
Collapse
Affiliation(s)
- An-Qi Yi
- Department of Hepatobiliary Surgery, Yanbian University Affiliated Hospital, Yanji 133099, Jilin Province, China
| | - Guang-Hua Xie
- Department of Hepatobiliary Surgery, Yanbian University Affiliated Hospital, Yanji 133099, Jilin Province, China
| |
Collapse
|
43
|
An P, Wang J, Fan R. Identifying and validating PLAU as a potential prognostic biomarker for PDAC. Sci Rep 2025; 15:12515. [PMID: 40216916 PMCID: PMC11992124 DOI: 10.1038/s41598-025-97629-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a prevalent cancer with a high mortality rate. This study aims to identify and validate biomarkers for early PDAC diagnosis. We employed the GEO2R online tool to screen differentially expressed genes (DEGs), construct protein interaction networks, and perform functional enrichment analysis, survival prognosis analysis, and expression level validation. We identified 260 DEGs, comprising 165 upregulated genes and 95 downregulated genes. Following functional enrichment and survival analysis, we selected plasminogen activator urokinase (PLAU) for the RNA and protein level verification and preliminary cell phenotype analysis. We found that PLAU knockdown inhibits the proliferation and survival of pancreatic cancer cells. Therefore, PLAU may serve as a potential biomarker, offering new strategies for understanding PDAC's pathological mechanisms.
Collapse
Affiliation(s)
- Peng An
- Material Evidence Technology Center, School of Law, Tianjin University of Commerce, Tianjin, 300134, China.
| | - Junhong Wang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Rong Fan
- Central Laboratory, Tianjin Xiqing hospital, Tianjin, 300380, PR China.
| |
Collapse
|
44
|
Wu C, Xie X, Yang X, Du M, Lin H, Huang J. Applications of gene pair methods in clinical research: advancing precision medicine. MOLECULAR BIOMEDICINE 2025; 6:22. [PMID: 40202606 PMCID: PMC11982013 DOI: 10.1186/s43556-025-00263-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: 09/06/2024] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
The rapid evolution of high-throughput sequencing technologies has revolutionized biomedical research, producing vast amounts of gene expression data that hold immense potential for biological discovery and clinical applications. Effectively mining these large-scale, high-dimensional data is crucial for facilitating disease detection, subtype differentiation, and understanding the molecular mechanisms underlying disease progression. However, the conventional paradigm of single-gene profiling, measuring absolute expression levels of individual genes, faces critical limitations in clinical implementation. These include vulnerability to batch effects and platform-dependent normalization requirements. In contrast, emerging approaches analyzing relative expression relationships between gene pairs demonstrate unique advantages. By focusing on binary comparisons of two genes' expression magnitudes, these methods inherently normalize experimental variations while capturing biologically stable interaction patterns. In this review, we systematically evaluate gene pair-based analytical frameworks. We classify eleven computational approaches into two fundamental categories: expression value-based methods quantifying differential expression patterns, and rank-based methods exploiting transcriptional ordering relationships. To bridge methodological development with practical implementation, we establish a reproducible analytical pipeline incorporating feature selection, classifier construction, and model evaluation modules using real-world benchmark datasets from pulmonary tuberculosis studies. These findings position gene pair analysis as a transformative paradigm for mining high-dimensional omics data, with direct implications for precision biomarker discovery and mechanistic studies of disease progression.
Collapse
Affiliation(s)
- Changchun Wu
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xueqin Xie
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xin Yang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Mengze Du
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China
| | - Hao Lin
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Jian Huang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| |
Collapse
|
45
|
Cao W, Sergeeva O, Julian W, Kresak A, Lusinger D, Schneider J, Berridge MS, Sexton S, Wojtylak P, Li Q, Liu W, Chan ER, Saunthararajah Y, Lee Z. PET imaging of hepatocellular carcinoma with [ 124I]IV-14. EJNMMI Res 2025; 15:35. [PMID: 40192905 PMCID: PMC11977070 DOI: 10.1186/s13550-025-01227-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/16/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Currently, positron emission tomography (PET) plays no clear role in clinical imaging and management of hepatocellular carcinoma (HCC). New radiotracers for new target(s) are needed for PET imaging of HCC. Uridine-cytidine kinase 2 (UCK2) is a rate-limiting enzyme of the pyrimidine salvage synthesis pathway to phosphorylate uridine and cytidine. Studies have demonstrated that UCK2 is overexpressed in many types of solid cancers including HCC and is associated with the poor prognosis and proliferation of HCC. This study reported PET imaging using a UCK2-specific radiotracer with a clinically relevant anima model of spontaneously occurring HCC in the woodchucks. METHODS This study used 3'-(E)-(2-iodovinyl) uridine (IV-14), which is derived from a UCK2-selective antitumor agent 3'-(Ethynyl)uridine (EUrd), a cytotoxic ribonucleoside analogs of uridine. By radiolabeling IV-14 with Iodine-124 (124I), a UCK2-specific radiotracer [124I]IV-14 was obtained for PET imaging of UCK2. A naturally occurring woodchuck model of HCC following chronic viral hepatitis infection was used for PET imaging. Potassium iodide (KI) was tested in one of the three animals to block possible uptake of free 124I from de-iodination of [124I]IV-14. RESULTS We confirmed that UCK2 expression is higher in the woodchuck model of HCC than in the surrounding hepatic tissue, similar to human UCK2 that is highly expressed in human HCC. PET imaging with [124I]IV-14 showed a strong uptake in woodchuck HCC with low background uptake at one-hour post-injection. De-iodination did not seem to be an issue for PET imaging. CONCLUSION Our results demonstrate that UCK2 is a viable target for imaging HCC and has the potential for targeted endoradiotherapy of HCC.
Collapse
Affiliation(s)
- Wei Cao
- Radiology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Olga Sergeeva
- Radiology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - William Julian
- Radiology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Adam Kresak
- Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | | | | | | | - Sandra Sexton
- Medical Oncology, Roswell Park Cancer Institute, Buffalo, NY, 14263, USA
| | - Patrick Wojtylak
- Nuclear Medicine, Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Qiubai Li
- Nuclear Medicine, Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Wendy Liu
- Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Ernest Ricky Chan
- Cleveland Institute for Computational Biology, Cleveland, OH, 44106, USA
| | | | - Zhenghong Lee
- Radiology, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Nuclear Medicine, Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA.
- Radiology and Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH, 44106, USA.
| |
Collapse
|
46
|
Napoli N, Ginesini M, Kauffmann EF, Barbarello L, Caniglia F, Costa F, Lombardo C, Perrone VG, Viti V, Amorese G, Boggi U. Navigating the learning curve of robotic pancreatoduodenectomy: Competency, proficiency, and mastery in a first-generation robotic surgeon with established open pancreatic expertise. Surgery 2025:109347. [PMID: 40180836 DOI: 10.1016/j.surg.2025.109347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 04/05/2025]
Abstract
OBJECTIVE This study delineates the learning curve of robotic pancreatoduodenectomy for first-generation surgeons, using the Comprehensive Complication Index to assess patient outcomes. BACKGROUND Robotic pancreatoduodenectomy is a promising alternative to open pancreatoduodenectomy, but patient safety and quality of outcomes during the learning phase remain critical. METHODS We retrospectively analyzed 313 consecutive robotic pancreatoduodenectomies performed by a single surgeon. The cumulative-sum method defined the learning curve, and Comprehensive Complication Index, adjusted for robotic pancreatoduodenectomy difficulty by PD-ROBOSCORE, was the dependent variable. RESULTS The median PD-ROBOSCORE was 8 (4.8-10.9), and the median Comprehensive Complication Index was 29.6 (20.9-39.5). At 90 days, severe morbidity and mortality rates were 24% and 5.4%, respectively. Three learning phases were identified: competency (63 procedures), proficiency (176), and mastery (263). Early phases involved simpler cases, whereas later phases showed greater complexity and a higher proportion of patients with ASA scores >2. Pancreatic cancer cases tripled in phases 2 and 3. Each phase showed progressive reductions in operative time and Comprehensive Complication Index. The mastery phase demonstrated further improvements in Comprehensive Complication Index, lymph node harvest, and margin status. Compared with proficiency, mastery saw improved outcomes in delayed gastric emptying, harvested lymph nodes, and R1 rates in pancreatic cancer. Operative time was longer, but morbidity and mortality remained stable. CONCLUSION The robotic pancreatoduodenectomy learning process involves competency, proficiency, and mastery phases. Structured training programs may accelerate this learning curve, but high procedural volumes are essential to improve outcomes. Future studies should account for surgeon experience and case complexity when evaluating robotic pancreatoduodenectomy outcomes.
Collapse
Affiliation(s)
- Niccolò Napoli
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | - Michael Ginesini
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | | | - Linda Barbarello
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | - Fabio Caniglia
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | - Francesca Costa
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | - Carlo Lombardo
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | | | - Virginia Viti
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | - Gabriella Amorese
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy; Division of Anesthesia and Intensive Care, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Ugo Boggi
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy.
| |
Collapse
|
47
|
Kaplan‐Arabaci O, Dančišinová Z, Paulsen RE. The Chicken Embryo: An Alternative Animal Model in Development, Disease and Pharmacological Treatment. Pharmacol Res Perspect 2025; 13:e70086. [PMID: 40113588 PMCID: PMC11925699 DOI: 10.1002/prp2.70086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/14/2025] [Accepted: 03/08/2025] [Indexed: 03/22/2025] Open
Abstract
To examine various medications and substances, in vivo models such as rats and mice are routinely used. However, it is utterly desirable to reduce extensive amounts of animals for these experimental models, which are costly and time-consuming. Animals are frequently put through a variety of procedures that could cause them pain, distress, or even harm; therefore, it is important to think about the ethical ramifications of using them in research. Thus, by following the three R's of animal research: reduction, replacement, and refinement, living animals used in studies should be minimized. The embryo of Gallus gallus, the domestic chicken, is a great model to research many different diseases and conditions. Its efficient blood supply from the chorioallantoic membrane gives us a unique possibility to administer chemicals or cells to the embryo in a noninvasive manner. In this review, we evaluate some advantages and disadvantages of using the developing chicken as an alternative in vivo model for development, disease, and pharmacological treatment. We focus on the top two leading causes of death: neurological disorders and cancer. We present a number of studies that describe the use of the chicken embryo in neuroscience and neurodevelopment research, in cancer research, and pharmacodynamic and pharmacokinetic studies. These studies show that the chicken embryo is an inexpensive, readily available, self-sufficient model with a short incubation period, high accessibility, and ideal for drug screening, making it an appealing model that can provide insightful biological and pharmacological information.
Collapse
Affiliation(s)
- Oykum Kaplan‐Arabaci
- Section for Pharmacology and Pharmaceutical Biosciences, Department of PharmacyFaculty of Mathematics and Natural Sciences, University of OsloOsloNorway
| | - Zuzana Dančišinová
- Section for Pharmacology and Pharmaceutical Biosciences, Department of PharmacyFaculty of Mathematics and Natural Sciences, University of OsloOsloNorway
- Institute of NeurobiologyBiomedical Research Center, Slovak Academy of SciencesKošiceSlovakia
| | - Ragnhild Elisabeth Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of PharmacyFaculty of Mathematics and Natural Sciences, University of OsloOsloNorway
| |
Collapse
|
48
|
Moosavi F, Firoozi R, Tavakkoli M, Nazari S, Alipour A, Firuzi O. Combination of chemotherapy and c-MET inhibitors has synergistic effects in c-MET overexpressing pancreatic cancer cells. Biochimie 2025; 231:73-83. [PMID: 39675659 DOI: 10.1016/j.biochi.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/17/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains as one of the most lethal malignancies. c-MET is an important oncogenic kinase involved in PDAC progression. We determined the anticancer effect of c-MET inhibitors, crizotinib and cabozantinib, combined with chemotherapeutic agents, doxorubicin, oxaliplatin and gemcitabine, against different PDAC and a lung adenocarcinoma cell line expressing different levels of c-MET. MTT assay was performed to assess cell growth inhibition. Synergistic combinations were evaluated in spheroid cultures, while apoptosis was determined through Hoechst33258 staining. The effect of drug combinations on cell cycle and apoptosis induction was examined by RNase/PI flow cytometric assay. We also evaluated reactive oxygen species (ROS) levels using 2',7'-dichlorofluorescein-diacetate (DCFH-DA) assay to explore the possible mechanisms contributing to synergism. Combination of crizotinib or cabozantinib with doxorubicin exhibited synergistic effects in c-MET overexpressing cells. Conversely, combinations of c-MET inhibitors with other agents were additive or even antagonistic. Combination index (CI) values calculated with Calcusyn software were 0.631-0.730 for crizotinib and 0.542-0.746 for cabozantinib co-administered with doxorubicin. These synergistic combinations showed significant spheroid growth inhibition and apoptosis induction in Suit-2, c-MET dependent PDAC cells. These combinations also significantly increased the number of cells in both apoptotic sub-G1 phase and the G2/M phase compared to single-drug treatment. Increased ROS production seemed to be a possible mechanism underlying synergism. In conclusion, c-MET inhibitors synergize with DNA damaging agent, doxorubicin, in cancer cells with c-MET overexpression, indicating that these combination therapies may be a promising cancer therapeutic strategy.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roya Firoozi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marjan Tavakkoli
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Nazari
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Alipour
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
49
|
Yang Z, Tang Y, Wu X, Wang J, Yao W. MicroRNA-130b Suppresses Malignant Behaviours and Inhibits the Activation of the PI3K/Akt Signaling Pathway by Targeting MET in Pancreatic Cancer. Biochem Genet 2025; 63:1660-1685. [PMID: 38607540 PMCID: PMC11929638 DOI: 10.1007/s10528-024-10696-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/10/2024] [Indexed: 04/13/2024]
Abstract
There has been interested in the microRNAs' roles in pancreatic cancer (PC) cell biology, particularly in regulating pathways related to tumorigenesis. The study aimed to explore the hub miRNAs in PC and underlying mechanisms by bioinformatics and fundamental experiments. RNA datasets collected from the Gene Expression Omnibus were analysed to find out differentially expressed RNAs (DERNAs). The miRNA-mRNA and protein-protein interaction (PPI) networks were built. The clinicopathological features and expressions of hub miRNAs and hub mRNAs were explored. Dual-luciferase reporter gene assay was performed to assess the interaction between microRNA and target gene. RT-qPCR and western blot were employed to explore RNA expression. The roles of RNA were detected by CCK-8 test, wound healing, transwell, and flow cytometry experiment. We verified 40 DEmiRNAs and 1613 DEmRNAs, then detected a total of 69 final functional mRNAs (FmRNAs) and 23 DEmiRNAs. In the miRNA-mRNA networks, microRNA-130b (miR-130b) was the hub RNA with highest degrees. Clinical analysis revealed that miR-130b was considerably lower expressed in cancerous tissues than in healthy ones, and patients with higher-expressed miR-130b had a better prognosis. Mechanically, miR-130b directly targeted MET in PC cells. Cell functional experiments verified that miR-130b suppressed cell proliferation, migration, promoted apoptosis, and inhibited the PI3K/Akt pathway by targeting MET in PC cells. Our findings illustrated the specific molecular mechanism of miR-130b regulating PC progress. The miR-130b/MET axis may be an alternative target in the therapeutic intervention of PC and provide an opportunity to deepen our understanding of the pathogenesis of PC.
Collapse
Affiliation(s)
- Zilin Yang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuming Tang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xuejiao Wu
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiancheng Wang
- Department of General Surgery, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Weiyan Yao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
50
|
Chandra S, Halfdanarson TR, Carlson EE, Rabe KG, Mahipal A, Majumder S, Bamlet WR, Horibe M, Tella SH, Shariq O, Carr RM, Cleary SP, Oberg AL, Antwi SO. Discordant risk factors between pancreatic neuroendocrine neoplasms and pancreatic ductal adenocarcinoma. Endocr Relat Cancer 2025; 32:e240142. [PMID: 39932006 PMCID: PMC11896652 DOI: 10.1530/erc-24-0142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/13/2025]
Abstract
Pancreatic neuroendocrine neoplasm (panNEN) is a rare malignancy and the second most common type of pancreatic cancer after pancreatic ductal adenocarcinoma (PDAC), but its etiology is poorly understood. We investigated whether the risk factors of panNEN are concordant with those known for PDAC. We performed the largest case-control study to date on panNENs, comprising 927 sporadic nonfunctional panNEN cases and 1807 frequency-matched controls, using data from the Mayo Clinic Biospecimen Resource for Pancreas Research. We assessed associations for obesity, first-degree family history of pancreatic cancer, cigarette smoking, overall type II diabetes mellitus (T2DM), new-onset T2DM (<1 year before panNEN diagnosis), longstanding T2DM (≥5 years), alcohol intake and aspirin use. Multivariable logistic regression was used to calculate odds ratios and 95% confidence intervals (CIs). Our results show that overall T2DM (OR = 1.71, 95% CI: 1.37-2.14) and new-onset T2DM (OR = 2.65, 95% CI: 1.92-3.69) are associated with higher odds of panNEN, but not longstanding T2DM (OR = 1.29, 95% CI: 0.94-1.75). A non-significant elevated odds of panNEN was observed among participants with a positive family history of pancreatic cancer (OR = 1.44, 95% CI: 0.96-2.14). Alcohol use was inversely related to panNEN (OR = 0.52, 95% CI: 0.42-0.66, ever-vs-never). No association was observed for smoking, obesity or aspirin use. These findings indicate that overall T2DM and new-onset T2DM are associated with higher odds of panNEN. Unlike PDAC, alcohol use was inversely related to panNEN, and we found no associations for cigarette smoking, obesity or aspirin use. These results indicate differences in the risk factor profiles of panNEN and PDAC.
Collapse
Affiliation(s)
- Shruti Chandra
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Thorvardur R Halfdanarson
- Division of Medical Oncology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Erin E Carlson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Kari G Rabe
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Amit Mahipal
- Department of Oncology, Seidman Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - William R Bamlet
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Masayasu Horibe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Sri Harsha Tella
- Department of Hematology and Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Omair Shariq
- Department of General Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ryan M Carr
- Division of Medical Oncology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Sean P Cleary
- Department of General Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Division of Hepatobiliary and Pancreatic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ann L Oberg
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Samuel O Antwi
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, USA
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|