1
|
Zucchini V, D'Acapito F, Rapposelli IG, Framarini M, Di Pietrantonio D, Turrini R, Pozzi E, Ercolani G. Impact of RAS, BRAF mutations and microsatellite status in peritoneal metastases from colorectal cancer treated with cytoreduction + HIPEC: scoping review. Int J Hyperthermia 2025; 42:2479527. [PMID: 40101749 DOI: 10.1080/02656736.2025.2479527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) has shown survival benefits in select patients with peritoneal metastases (PM) from colorectal cancer (CRC). Molecular alterations, particularly RAS/BRAF mutations and Microsatellite Instability (MSI), play crucial roles in prognostic stratification and treatment planning, influencing both disease-free survival (DFS) and overall survival (OS). This scoping review evaluates the prognostic role of MSI and RAS/BRAF mutations in patients with PM-CRC treated with CRS-HIPEC. DESIGN A literature search was conducted across several databases to identify papers published between 2000 and September 2024. We selected 18 publications that considered DFS and OS as primary or secondary outcomes in patients with RAS/BRAF mutations and MSI following CRS-HIPEC treatment. Studies involving appendiceal cancer, peritoneal disease from non-CRC, pediatric patients, or subjects not treated with CRS-HIPEC were excluded. RESULTS Most studies suggest that RAS and BRAF mutations have a negative influence on survival outcomes. While inconsistencies exist, RAS mutations are generally associated with worse DFS. Specific KRAS subtypes such as KRASMUT2 or KRAS G12V and the BRAF V600 variant correlate with poorer prognosis. MSI status appears to attenuate the adverse effects of RAS/BRAF mutations on survival, although conflicting data persist. CONCLUSION RAS and BRAF mutations correlate with poorer outcomes in PM-CRC, underscoring the need for mutation-informed strategies to refine HIPEC and systemic therapies. Recognizing subtypes may improve patient selection for CRS-HIPEC, optimizing both local disease control and long-term survival. Future research should incorporate these molecular profiles to enhance therapeutic decision-making and better address this challenging condition.
Collapse
Affiliation(s)
- Valentina Zucchini
- Department of Medical and Surgical Sciences - DIMEC, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Fabrizio D'Acapito
- General and Oncologic Department of Surgery, Morgagni-Pierantoni Hospital, AUSL Romagna, Forlì, Italy
| | - Ilario Giovanni Rapposelli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Massimo Framarini
- General and Oncologic Department of Surgery, Morgagni-Pierantoni Hospital, AUSL Romagna, Forlì, Italy
| | - Daniela Di Pietrantonio
- General and Oncologic Department of Surgery, Morgagni-Pierantoni Hospital, AUSL Romagna, Forlì, Italy
| | - Riccardo Turrini
- Department of Medical and Surgical Sciences - DIMEC, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Eleonora Pozzi
- Department of Medical and Surgical Sciences - DIMEC, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Giorgio Ercolani
- Department of Medical and Surgical Sciences - DIMEC, Alma Mater Studiorum - University of Bologna, Bologna, Italy
- General and Oncologic Department of Surgery, Morgagni-Pierantoni Hospital, AUSL Romagna, Forlì, Italy
| |
Collapse
|
2
|
Zhang J, Li G, Wu R, Shi L, Tian C, Jiang H, Che H, Jiang Y, Jin Z, Yu R, Liu X, Zhang X. The m6A RNA demethylase FTO promotes radioresistance and stemness maintenance of glioma stem cells. Cell Signal 2025; 132:111782. [PMID: 40185350 DOI: 10.1016/j.cellsig.2025.111782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 03/09/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Glioblastoma (GBM) was the most common and deadliest malignant brain tumor in adults, with a poor prognosis. Effective targeted drugs are still lacking, and the presence of glioblastoma stem cells (GSC) is a major factor contributing to radiotherapy resistance. Screening for targeted drugs that can sensitize GBM to radiotherapy is crucial. FTO is considered an attractive potential target for tumor therapy, as it mediates m6A demethylation to regulate the stability of target genes. In this study, we evaluated the role of FTO inhibition in promoting the sensitivity of GSC cells to radiotherapy through tumor sphere formation assays, cell apoptosis assays, and in situ GSC tumor models. We preliminarily explored the molecular mechanisms by transcriptome sequencing and m6A methylation sequencing to investigate how inhibiting FTO increases radiotherapy sensitivity. The results showed that downregulation of FTO expression or FTO inhibitor FB23-2 combined with radiotherapy significantly inhibited GSC cell proliferation and self-renewal and increased apoptosis. FB23-2 combined with radiotherapy effectively inhibited intracranial tumor growth in mice and prolonged the survival of tumor-bearing mice. Furthermore, FTO inhibition sustained the increase of γH2AX expression induced by radiotherapy while decreasing Rad51 expression. Importantly, we found that inhibiting FTO could increase m6A methylation modification of VEGFA, thereby downregulating both mRNA and protein expression of VEGFA. Our findings provide a new therapeutic strategy for enhancing GBM radiotherapy sensitivity and lay the theoretical and experimental groundwork for clinical trials targeting FTO.
Collapse
Affiliation(s)
- Junhao Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang 453003, Henan, China
| | - Guoxi Li
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Runqiu Wu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin Shi
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of General Surgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cong Tian
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongyan Jiang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongyu Che
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yongang Jiang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhiyong Jin
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rutong Yu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Xuejiao Liu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Xu Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
3
|
Chaturvedi B, Khandare M, Mishra D, Yadav SK, Agarwal P, Sharma D. Optimising Axillary Staging in Resource-Constrained Settings: A Prospective Validation of Axillary Ultrasound and Touch Imprint Cytology in Predicting Pathologically Negative Axillae in cT2-3 Breast Cancer. Cytopathology 2025; 36:386-390. [PMID: 40151007 DOI: 10.1111/cyt.13484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Management of axillary lymph nodes (ALNs) in breast cancer patients remains pivotal for staging and planning therapeutic strategies. However, In low-resource settings, achieving accurate axillary staging while avoiding overtreatment remains a challenge as the majority of patients present with advanced stage. In this prospective validation study, we assessed the efficacy of axillary ultrasound (AUS) combined with touch imprint cytology (TIC) for predicting negative axillary status in cT2-3 breast cancer patients. METHODS This study was a prospective, single-centre validation study conducted in the Breast and Endocrine Unit of the Department of Surgery and the Department of Pathology in a tertiary teaching hospital in central India from September 2022 to April 2024. Eligible participants included adult female patients (aged ≥ 18 years) with core needle biopsy-proven invasive breast cancer classified as cT2-3, cN0, and scheduled for primary surgical treatment. The primary outcomes were the Sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV) and accuracy of the AUS + TIC approach in predicting pathologically negative axillary status. RESULTS AUS + TIC had a sensitivity of 100% (95% CI: 47.82%-100%), a specificity of 100% (95% CI: 91.19%-100%) and an overall accuracy of 100% (95% CI: 92.13%-100%). There were no false negatives. CONCLUSION Our findings suggest that the combination of AUS + TIC provides a reliable technique with high diagnostic accuracy, sensitivity, and specificity for assessing ALN in low resource settings.
Collapse
Affiliation(s)
- Balmik Chaturvedi
- Department of Pathology, NSCB Medical College, Jabalpur, Madhya Pradesh, India
| | - Muktesh Khandare
- Department of Pathology, NSCB Medical College, Jabalpur, Madhya Pradesh, India
| | - Devashish Mishra
- Department of Radiodiagnosis, NSCB Medical College, Jabalpur, Madhya Pradesh, India
| | - Sanjay Kumar Yadav
- Department of Surgery, NSCB Medical College, Jabalpur, Madhya Pradesh, India
| | - Pawan Agarwal
- Department of Surgery, NSCB Medical College, Jabalpur, Madhya Pradesh, India
| | - Dhananjaya Sharma
- Department of Surgery, NSCB Medical College, Jabalpur, Madhya Pradesh, India
| |
Collapse
|
4
|
Qiu X, Gao Q, Wang J, Zhang Z, Tao L. The microbiota-m 6A-metabolism axis: Implications for therapeutic strategies in gastrointestinal cancers. Biochim Biophys Acta Rev Cancer 2025; 1880:189317. [PMID: 40222422 DOI: 10.1016/j.bbcan.2025.189317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
Gastrointestinal (GI) cancers remain a leading cause of cancer-related mortality worldwide, with metabolic reprogramming recognized as a central driver of tumor progression and therapeutic resistance. Among the key regulatory layers, N6-methyladenosine (m6A) RNA modification-mediated by methyltransferases (writers such as METTL3/14), RNA-binding proteins (readers like YTHDFs and IGF2BPs), and demethylases (erasers including FTO and ALKBH5), plays a pivotal role in controlling gene expression and metabolic flux in the tumor context. Concurrently, the gut microbiota profoundly influences GI tumorigenesis and immune evasion by modulating metabolite availability and remodeling the tumor microenvironment. Recent evidence has uncovered a bidirectional crosstalk between microbial metabolites and m6A methylation: microbiota-derived signals dynamically regulate m6A deposition on metabolic and immune transcripts, while m6A modifications, in turn, regulate the stability and translation of key mRNAs such as PD-L1 and FOXP3. This reciprocal interaction forms self-reinforcing epigenetic circuits that drive tumor plasticity, immune escape, and metabolic adaptation. In this review, we dissect the molecular underpinnings of the microbiota-m6A-metabolism axis in GI cancers and explore its potential to inform novel strategies in immunotherapy, metabolic intervention, and microbiome-guided precision oncology.
Collapse
Affiliation(s)
- Xiuxiu Qiu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qi Gao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jiahui Wang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhanxia Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Li Tao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
5
|
Qiu TH, Wen HY, Huang YL. Significance of hemoglobin and hematocrit changes in predicting patient survival and efficacy of neoadjuvant chemotherapy for advanced gastric cancer. World J Gastrointest Oncol 2025; 17:104592. [DOI: 10.4251/wjgo.v17.i6.104592] [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: 03/07/2025] [Revised: 03/31/2025] [Accepted: 04/24/2025] [Indexed: 06/13/2025] Open
Abstract
BACKGROUND Advanced gastric cancer is characterized by fast tumor growth and aggressive biological behavior. During neoadjuvant chemotherapy, patients are at risk of distant metastasis or local progression. Anemia is a frequent complication in these patients.
AIM To analyze whether changes in hemoglobin and hematocrit can predict the survival and efficacy of neoadjuvant chemotherapy in patients with advanced gastric cancer.
METHODS The clinical data of 185 patients with advanced gastric cancer admitted to the Third Affiliated Hospital of Chengdu Medical College, Pidu District People’s Hospital, Chengdu, China, between January 2016 and January 2021, were retrospectively analyzed. All patients underwent a tegafur + oxaliplatin + apatinib chemotherapy regimen. According to the efficacy of chemotherapy, they were divided into an effective group (complete or partial response, n = 121) and an ineffective group (stable disease or disease progression, n = 64). The factors related to chemotherapy efficacy in patients with advanced gastric cancer were analyzed by univariate and logistic multivariate analyses. The 3-year survival rates of the patients with different hemoglobin and hematocrit levels were compared.
RESULTS Univariate analysis showed that the proportion of patients with a tumor diameter > 5 cm, non-tubular adenocarcinoma, lymph node metastasis, hematocrit < 33%, low mean red blood cell (RBC) protein content, low RBC distribution width, hemoglobin < 107 g/L, and platelets > 266 × 109/L in the ineffective group were significantly higher than those in the effective group (P < 0.05). Logistic multivariate analysis showed that a tumor diameter > 5 cm, lymph node metastasis, ≤ 3 chemotherapy cycles, hematocrit < 33%, and hemoglobin < 107 g/L are risk factors for neoadjuvant chemotherapy failure in advanced gastric cancer (P < 0.05). The 1-year, 2-year, and 3-year survival rates in the effective group were 93.39%, 83.47%, and 60.33%, respectively. These rates were significantly higher than those in the ineffective group (P < 0.05). The 1-year, 2-year, and 3-year survival rates of patients with hematocrit < 33% were 74.67%, 49.33%, and 29.33%, respectively, which were significantly lower than those of patients with hematocrit ≥ 33% (P < 0.05). The 1-year, 2-year, and 3-year survival rates of patients with hemoglobin < 107 g/L were 80.39%, 58.82%, and 39.22%, respectively, which were significantly lower than those of patients with hemoglobin ≥ 107 g/L (P < 0.05).
CONCLUSION Hematocrit < 33% and hemoglobin < 107 g/L are risk factors for chemotherapy failure in patients with advanced gastric cancer. They are associated with poorer prognosis and reduced 3-year survival rates.
Collapse
Affiliation(s)
- Ti-Hong Qiu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu 611730, Sichuan Province, China
| | - Hong-You Wen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu 611730, Sichuan Province, China
| | - Yi-Long Huang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu 611730, Sichuan Province, China
| |
Collapse
|
6
|
Ren Q, Luo X, Yu C, Xiang Y, Wu D, Zhao Y, Li Z. Mechanisms of Jisheng-Wumei tablet in laryngeal cancer treatment: a network pharmacology analysis of baicalein's targeting of cytochrome C for laryngeal cancer inhibition. Discov Oncol 2025; 16:1027. [PMID: 40481938 PMCID: PMC12145384 DOI: 10.1007/s12672-025-02829-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
OBJECTIVES Jisheng-Wumei Tablet (JSWMT) is traditionally used in Chinese medicine for cancer therapy and shows potential efficacy against laryngeal cancer. This study aimed to elucidate the active components of JSWMT, with a specific focus on the role of baicalein in targeting cytochrome C (CYCS) to inhibit laryngeal cancer. METHODS Utilizing network pharmacology, we identified the active ingredients of JSWMT and their corresponding drug targets by leveraging data from the TCMSP and GeneCards databases to pinpoint laryngeal cancer-related targets. Key targets were identified through survival analysis of TCGA expression data, and molecular docking was employed to evaluate the binding affinity between JSWMT compounds and these targets. RESULTS Our research identified 35 effective JSWMT ingredients that interact with 132 drug targets, which intersect with 421 targets related to laryngeal cancer. Survival analysis highlighted IL2, CYCS, and CTNNB1 as critical targets significantly correlated with patient survival. Molecular docking demonstrated a strong affinity between CYCS and baicalein, indicating that baicalein is JSWMT's principal active component against laryngeal cancer. CONCLUSION Baicalein is identified as the key active ingredient in JSWMT, targeting CYCS to inhibit the growth of laryngeal cancer cells. This investigation highlights baicalein's potential mechanism of action and underscores the importance of integrating traditional Chinese medicine with contemporary cancer treatment strategies, offering a novel approach to laryngeal cancer therapy.
Collapse
Affiliation(s)
- Quanwei Ren
- Chongqing Traditional Chinese Medicine Hospital, No. 6 Panxi-7th Road, Jiangbei District, Chongqing, 400021, China
| | - Xiaoli Luo
- Chongqing Traditional Chinese Medicine Hospital, No. 6 Panxi-7th Road, Jiangbei District, Chongqing, 400021, China
| | - Chao Yu
- Chongqing Traditional Chinese Medicine Hospital, No. 6 Panxi-7th Road, Jiangbei District, Chongqing, 400021, China
| | - Yilin Xiang
- Chongqing Traditional Chinese Medicine Hospital, No. 6 Panxi-7th Road, Jiangbei District, Chongqing, 400021, China
| | - Di Wu
- Chongqing Traditional Chinese Medicine Hospital, No. 6 Panxi-7th Road, Jiangbei District, Chongqing, 400021, China
| | - Yanli Zhao
- Chongqing Traditional Chinese Medicine Hospital, No. 6 Panxi-7th Road, Jiangbei District, Chongqing, 400021, China.
| | - Zhongwan Li
- Chongqing General Hospital, No.312 Zhongshan-1 Road, Yuzhong District, Chongqing, 401147, China.
| |
Collapse
|
7
|
Li Y, Xue J, Ma Y, Ye K, Zhao X, Ge F, Zheng F, Liu L, Gao X, Wang D, Xia Q. The complex roles of m 6 A modifications in neural stem cell proliferation, differentiation, and self-renewal and implications for memory and neurodegenerative diseases. Neural Regen Res 2025; 20:1582-1598. [PMID: 38845217 PMCID: PMC11688559 DOI: 10.4103/nrr.nrr-d-23-01872] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/19/2024] [Accepted: 03/25/2024] [Indexed: 08/07/2024] Open
Abstract
N6-methyladenosine (m 6 A), the most prevalent and conserved RNA modification in eukaryotic cells, profoundly influences virtually all aspects of mRNA metabolism. mRNA plays crucial roles in neural stem cell genesis and neural regeneration, where it is highly concentrated and actively involved in these processes. Changes in m 6 A modification levels and the expression levels of related enzymatic proteins can lead to neurological dysfunction and contribute to the development of neurological diseases. Furthermore, the proliferation and differentiation of neural stem cells, as well as nerve regeneration, are intimately linked to memory function and neurodegenerative diseases. This paper presents a comprehensive review of the roles of m 6 A in neural stem cell proliferation, differentiation, and self-renewal, as well as its implications in memory and neurodegenerative diseases. m 6 A has demonstrated divergent effects on the proliferation and differentiation of neural stem cells. These observed contradictions may arise from the time-specific nature of m 6 A and its differential impact on neural stem cells across various stages of development. Similarly, the diverse effects of m 6 A on distinct types of memory could be attributed to the involvement of specific brain regions in memory formation and recall. Inconsistencies in m 6 A levels across different models of neurodegenerative disease, particularly Alzheimer's disease and Parkinson's disease, suggest that these disparities are linked to variations in the affected brain regions. Notably, the opposing changes in m 6 A levels observed in Parkinson's disease models exposed to manganese compared to normal Parkinson's disease models further underscore the complexity of m 6 A's role in neurodegenerative processes. The roles of m 6 A in neural stem cell proliferation, differentiation, and self-renewal, and its implications in memory and neurodegenerative diseases, appear contradictory. These inconsistencies may be attributed to the time-specific nature of m 6 A and its varying effects on distinct brain regions and in different environments.
Collapse
Affiliation(s)
- Yanxi Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jing Xue
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yuejia Ma
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ke Ye
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xue Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Fangliang Ge
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Feifei Zheng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lulu Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- Basic Medical Institute, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
- Key Laboratory of Heilongjiang Province for Genetically Modified Animals, Harbin Medical University, Harbin, Heilongjiang Province, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Dayong Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Qing Xia
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
8
|
Qin J, Chen B, Sun YH, Wang XX, Wu C, Zhang C. The predictive value of miR-132-3p combined with Prognostic Nutritional Index (PNI) for gastric cancer prognosis. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2025; 117:303-312. [PMID: 39784731 DOI: 10.17235/reed.2024.10882/2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
BACKGROUND previous studies have demonstrated that the Prognostic Nutritional Index (PNI) can predict the prognosis of gastric cancer (GC) patients. However, few studies have focused on the auxiliary role of miRNA in predicting the prognosis of GC. OBJECTIVE this study aimed to clarify the role of the combined use of miR-132-3p and PNI to predict the prognosis of GC patients. METHODS a total of 126 patients with GC who underwent surgical treatment at our institution were included in the study. The qRT-PCR data was used to assess the expression of miR-132-3p in tumor and adjacent normal tissues. The predictive value of miR-132-3p and PNI for postoperative prognosis, and the relationships between miR-132-3p, PNI, and preoperative clinical characteristics, were assessed using ROC, χ², Kaplan-Meier survival analysis, and Cox regression analysis. Bioinformatics analyses and dual-luciferase reporter assay were conducted to elucidate the biological function of miR-132-3p and the underlying molecular mechanisms. RESULTS miR-132-3p was downregulated in GC tumor tissues and significantly and positively correlated with PNI. Both miR-132-3p and PNI were significantly associated with TNM stage and lymph node metastasis. Postoperative GC patients with low miR-132-3p expression and low PNI had lower survival rates, and both were independent risk factors for poor prognosis. The combination of miR-132-3p and PNI demonstrated better sensitivity and specificity to predict postoperative prognosis than either indicator alone. miR-132-3p may be involved in DNA damage response, angiogenesis, and immune response associated with GC development. MAPK1 was identified as a target gene regulated by miR-132-3p in GC. CONCLUSION the combination of miR-132-3p and PNI can effectively improve the predictive value of postoperative prognosis in GC patients. The combination of miR-132-3p and PNI, which are strongly correlated with pathological characteristics and can be readily obtained, has a potential clinical applicability in the prognosis of patients with GC.
Collapse
Affiliation(s)
| | - Bixia Chen
- Gastroenterology, Jiangmen Central Hospital
| | - Yan-Hui Sun
- Nutrition, The Seventh Medical Center of Chinese PLA General Hospital
| | - Xiao-Xiao Wang
- Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital
| | - Cong Wu
- Nutrition, The Seventh Medical Center of Chinese PLA General Hospital, China
| | - Caihua Zhang
- Oncology, People's Hospital Affiliated to Chongqing Three Gorges Medical College
| |
Collapse
|
9
|
Tuo JY, Shen QM, Li ZY, Yang DN, Zou YX, Tan YT, Li HL, Xiang YB. Adherence to dietary guidelines and liver cancer risk: Results from two prospective cohort studies. Clin Nutr ESPEN 2025; 67:599-611. [PMID: 40187735 DOI: 10.1016/j.clnesp.2025.03.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND AND AIMS Although dietary factors have been extensively investigated as potential risk factors for liver cancer, the evidence is inconclusive. Our study systematically assessed the associations between ten dietary guidelines compliance scores and liver cancer risk among adult people, and found out the dietary patterns for liver cancer prevention. METHODS Participants of 59,844 men and 72,680 women, aged 40-74 years and living in urban Shanghai, were included in two prospective cohort studies conducted between 2002-2006 and 1996-2000, respectively. Dietary intakes were assessed during baseline in-person interviews using validated food-frequency questionnaires, and dietary guideline compliance scores were calculated by adjusting for total energy intake and adapting existing dietary recommendations. Hazards ratios (HRs) with 95 % confidence intervals (CIs) were evaluated by both tertile categories and per standard deviation (SD) increment using Cox proportional hazard regression models to assess the associations between ten dietary guideline compliance scores and liver cancer risk. RESULTS In the two cohorts, 431 male and 256 female incident liver cancer cases were identified during a mean follow-up of 11.90 and 17.44 years, respectively. There were no statistically significant associations between these ten dietary guidelines and male liver cancer risk (P > 0.05). In contrast, only the modified reversed Empirical Dietary Inflammation Pattern (rEDIP) tended to be associated with the low risk of female liver cancer (HR T3 vs. T1 = 0.67, 95 % CI: 0.48-0.92, Ptrend = 0.016, HR per SD = 0.94, 95 % CI: 0.85-1.03). The inverse associations appeared stronger between rEDIP and liver cancer risk at younger ages (<55 years) in women (HR per SD = 0.91, 95 % CI: 0.84-0.99) compared to the older women (≥55 years). There were suggestive but non-significant inverse associations between the modified Diabetes Risk Reduction Diet (mDRRD) (men: HR per SD = 0.92, 95 % CI: 0.84-1.02; women: HR per SD = 0.97, 95 % CI: 0.84-1.02) and the modified World Cancer Research Fund/American Institute for Cancer Research (mWCRF/AICR) (men: HR per SD = 0.93, 95 % CI: 0.84-1.02; women: HR per SD = 0.91, 95 % CI: 0.80-1.03) and liver cancer incidence. The associations of mDRRD (HR per SD = 0.82, 95 % CI: 0.75-0.98) and mWCRF/AICR (HR per SD = 0.83, 95 % CI: 0.74-0.99) on liver cancer risk were significant in men who ever smoked. CONCLUSIONS Our findings confirm that greater adherence to some healthy dietary patterns (i.e. rEDIP, mDRRD and mWCRF/AICR) is inversely associated with liver cancer risk, especially in certain populations. Future studies are required to confirm these findings and elucidate potential mechanisms.
Collapse
Affiliation(s)
- Jia-Yi Tuo
- State Key Laboratory of Systems Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Qiu-Ming Shen
- State Key Laboratory of Systems Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhuo-Ying Li
- State Key Laboratory of Systems Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Dan-Ni Yang
- State Key Laboratory of Systems Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Yi-Xin Zou
- State Key Laboratory of Systems Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; School of Public Health, Fudan University, Shanghai, PR China
| | - Yu-Ting Tan
- State Key Laboratory of Systems Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hong-Lan Li
- State Key Laboratory of Systems Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yong-Bing Xiang
- State Key Laboratory of Systems Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, PR China; School of Public Health, Fudan University, Shanghai, PR China.
| |
Collapse
|
10
|
Liang X, Yin S, Hu C, Tang D, Luo G, Liu Z. METTL14 Promotes Ischemic Stroke-induced Brain Injury by Stabilizing HDAC3 Expression in an m6A-IGF2BP3 Mechanism. Cell Biochem Biophys 2025; 83:1897-1907. [PMID: 39448421 DOI: 10.1007/s12013-024-01596-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 10/26/2024]
Abstract
N6-methyladenosine (m6A) modification is the most abundant post-transcriptional modification of mRNAs and has been identified to play critical roles in ischemic stroke (IS). Herein, this study aimed to investigate the function and mechanism of Methyltransferase-like 14 (METTL14) methylase in cerebral IS. Murine BV-2 microglial cell OGD/R models and rat middle cerebral artery occlusion (MCAO) models were established to mimic IS-induced neuronal damage in vitro and brain injury in vivo. Levels of METTL14, Histone Deacetylase 3 (HDAC3) and cGAS-STING axis-related proteins were detected using qRT-PCR or western blotting. Cell proliferation and inflammation were assessed by CCK-8 assay, EdU assay and ELISA. Flow cytometry detected microglia polarization. Cell pyroptosis was analyzed by detecting related-protein markers by western blotting. The m6A modification was determined by methylated RNA immunoprecipitation assay. Brain injury was analyzed by evaluating infarct volume and neurologic score. METTL14 levels were higher in OGD/R-induced microglial cells, primary microglia and infarct brain tissues of rat MCAO models. Functionally, METTL14 silencing reversed OGD/R-induced proliferation inhibition, inflammation and pyroptosis in microglial cells and primary microglia in vitro, and ameliorated cerebral ischemic injury in rat MCAO models. Mechanistically, METTL14 induced HDAC3 m6A modification in an IGF2BP3-dependent manner, and could activate cGAS-STING pathway through HDAC3. Moreover, HDAC3 overexpression reversed the neuroprotective effects of METTL14 silencing. METTL14 silencing reversed ischemic stroke-induced brain injury by inducing HDAC3 m6A modification in an IGF2BP3-dependent mechanism, recommending a novel insight for ameliorating cerebral ischemic stroke.
Collapse
Affiliation(s)
- Xuelin Liang
- The Medical Department of Neurology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Songhe Yin
- The Medical Department of Neurology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Canfang Hu
- The Medical Department of Neurology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Dingzhong Tang
- The Medical Department of Neurology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Guojun Luo
- The Medical Department of Neurology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Zhen Liu
- The Medical Department of Neurology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
11
|
Lu YM, Jin P, Wang HK, Shao XX, Hu HT, Jiang YJ, Li WY, Tian YT. Proximal gastrectomy with tubular stomach reconstruction vs total gastrectomy for proximal gastric cancer following neoadjuvant chemotherapy: A multicenter retrospective study. World J Gastrointest Surg 2025; 17:107579. [DOI: 10.4240/wjgs.v17.i5.107579] [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: 03/26/2025] [Revised: 04/08/2025] [Accepted: 04/25/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a major global health challenge, and the treatment of proximal GC in particular presents unique clinical and surgical complexities. Currently, there is no consensus on whether proximal gastrectomy (PG) or total gastrectomy (TG) should be used for advanced proximal GC, and the choice of postoperative gastrointestinal reconstruction method remains controversial.
AIM To compare the short-term efficacy, long-term survival, and postoperative reflux outcomes of PG with tubular stomach reconstruction vs TG with Roux-en-Y reconstruction in patients with proximal GC following neoadjuvant chemotherapy (NACT) in an effort to provide valuable insights for clinical decision-making regarding the optimal surgical approach.
METHODS A multicenter retrospective cohort study was conducted at two Chinese medical centers between December, 2012 and December, 2022. Patients with histologically confirmed proximal GC who received NACT followed by either PG with tubular stomach reconstruction or TG with Roux-en-Y reconstruction were included. Propensity score matching (PSM) was performed to balance baseline characteristics, and the primary endpoint was 5-year overall survival (OS). Secondary endpoints included recurrence-free survival (RFS), postoperative complications, and reflux severity.
RESULTS After PSM, 244 patients (122 PG, 122 TG) were finally included and all baseline characteristics were comparable between groups. The PG group had a significantly shorter operation time compared to the TG group (189.50 vs 215.00 minutes, P < 0.001), with no differences in intraoperative blood loss or postoperative complications (19.68% vs 14.75%, P = 0.792). The 5-year OS rates were 52.7% vs 45.5% (P = 0.330), and 5-year RFS rates were 54.3% vs 47.6% (P = 0.356) for the PG and TG groups, respectively. Reflux symptoms (18.0% vs 31.1%, P = 0.017) and clinically significant reflux based on gastroesophageal reflux disease questionnaire scores ≥ 8 (7.4% vs 21.3%, P < 0.001) were significantly less frequent in the PG group. Multivariate analysis identified histological differentiation (HR = 2.98, 95%CI: 2.03-4.36, P < 0.001) and tumor size (HR = 0.26, 95%CI: 0.17-0.41 for tumors ≤ 4 cm, P < 0.001) as independent prognostic factors.
CONCLUSION PG with tubular stomach reconstruction is comparable to TG in terms of surgical safety and long-term oncological outcomes for proximal GC patients following NACT. Additionally, PG has the advantages of shorter operation time and lower rates of postoperative reflux, suggesting potential benefits for patient quality of life. Notably, the analysis of postoperative prognostic factors, including histological differentiation and tumor size, further informs clinical decision-making and highlights the importance of individualized treatment strategies.
Collapse
Affiliation(s)
- Yi-Ming Lu
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Peng Jin
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Hai-Kuo Wang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xin-Xin Shao
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hai-Tao Hu
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu-Juan Jiang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wang-Yao Li
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan-Tao Tian
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
12
|
Sun Z, Qi T, Cheng B, Guo Y, Atehli D, Cui SW, Kang J, Guo Q. The Protective Effects of Pectic Polysaccharides on Dextran Sulfate Sodium-Induced Colitis in Drosophila melanogaster and Their Structure-Function Relationships. Nutrients 2025; 17:1738. [PMID: 40431477 PMCID: PMC12113875 DOI: 10.3390/nu17101738] [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: 04/15/2025] [Revised: 05/08/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Pectic polysaccharides exhibit therapeutic potential against intestinal inflammation. However, the influence of structural variations on their efficacy remains largely unexplored. METHODS This study investigated the structural and anti-inflammatory relationships of okra pectin (OP), citrus pectin (CP), apple pectin (AP), and hawthorn pectin (HP). Based on FT-IR spectra, CP was identified as a high-methoxyl pectin, with a degree of methyl esterification (DM) of 72.07 ± 3.86%. OP, AP, and HP were low-methoxyl pectins with the following DM values: 19.34 ± 3.04%, 32.11 ± 1.71%, and 38.67 ± 2.75%, respectively. RESULTS Monosaccharide composition analysis revealed that OP exhibited the highest abundance of RG-I regions among all the samples. Homogalacturonan (HG) was the predominant structural region in AP and HP, while CP contained both of the aforementioned structural regions. Our findings demonstrated that OP and CP significantly ameliorated dextran sulfate sodium (DSS)-induced colitis in the wild-type Drosophila melanogaster strain w1118, as evidenced by improved intestinal morphology, reinforced intestinal barrier function, and enhanced locomotor and metabolic activity. These effects were mediated by the inhibition of JAK/STAT signaling and the activation of the Nrf2/Keap1 pathway. Notably, reducing the molecular weight of CP to 18.18 kDa significantly enhanced its therapeutic efficacy, whereas a reduction in OP molecular weight to 119.12 kDa extended its median lifespan. CONCLUSIONS These findings first suggest that abundant RG-I structures and low molecular weight endowed pectins with significant anti-inflammatory activity.
Collapse
Affiliation(s)
- Zhenou Sun
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China; (Z.S.); (T.Q.); (B.C.); (Y.G.); (D.A.); (Q.G.)
| | - Tianyu Qi
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China; (Z.S.); (T.Q.); (B.C.); (Y.G.); (D.A.); (Q.G.)
| | - Boyu Cheng
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China; (Z.S.); (T.Q.); (B.C.); (Y.G.); (D.A.); (Q.G.)
| | - Yingxiao Guo
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China; (Z.S.); (T.Q.); (B.C.); (Y.G.); (D.A.); (Q.G.)
| | - Dima Atehli
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China; (Z.S.); (T.Q.); (B.C.); (Y.G.); (D.A.); (Q.G.)
| | - Steve W. Cui
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON N1G 5C9, Canada;
| | - Ji Kang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China; (Z.S.); (T.Q.); (B.C.); (Y.G.); (D.A.); (Q.G.)
| | - Qingbin Guo
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science and Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China; (Z.S.); (T.Q.); (B.C.); (Y.G.); (D.A.); (Q.G.)
| |
Collapse
|
13
|
Sakthi Velu K, Aslam M, Srinivasan R, Somu P, Mohandoss S. Flufenamic Acid-Loaded Electrospun Nanofibers Based on Chitosan/Poly(vinyl alcohol) Polymeric Composites for Drug Delivery in Biomedical Applications. Polymers (Basel) 2025; 17:1411. [PMID: 40430707 PMCID: PMC12114851 DOI: 10.3390/polym17101411] [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: 04/04/2025] [Revised: 05/15/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Nanostructured drug-delivery systems with enhanced therapeutic potential have gained attention in biomedical applications. Here, flufenamic acid (FFA)-loaded chitosan/poly(vinyl alcohol) (CHS/PVA; CSPA)-based electrospun nanofibers were fabricated and characterized for antibacterial, anticancer, and antioxidant activities. The FFA-loaded CSPA (FCSPA) nanofibers were characterized by scanning electron microscopy, Fourier-transform infrared spectroscopy, X-ray diffraction (XRD), and differential scanning calorimetry to evaluate their formation process, functional group interactions, and crystallinity. Notably, the average diameter of FCSPA nanofibers decreased with increasing CSPA contents (CSPA-1 to CSPA-3), indicating that FFA addition to CSPA-3 significantly decreased its diameter. Additionally, XRD confirmed the dispersion of FFA within the CSPA amorphous matrix, enhancing drug stability. FCSPA nanofibers exhibited a high swelling ratio (significantly higher than that of the CSPA samples). Biodegradation studies revealed that FCSPA exhibited accelerated weight loss after 72 h, indicating its improved degradation compared with those of other formulations. Furthermore, it exhibited a significantly high drug-encapsulation efficiency, ensuring sustained release. FCSPA nanofibers exhibited excellent antibacterial activity, inhibiting Staphylococcus aureus and Escherichia coli. Regarding anticancer activity, FCSPA decreased HCT-116 cell viability, highlighting its controlled drug-delivery potential. Moreover, FCSPA demonstrated superior antioxidation, scavenging DPPH free radicals. These findings highlight FCSPA nanofibers as multifunctional platforms with wound-healing, drug-delivery, and tissue-engineering potential.
Collapse
Affiliation(s)
- Kuppu Sakthi Velu
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea; (K.S.V.); (M.A.)
| | - Mohammad Aslam
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea; (K.S.V.); (M.A.)
| | - Ramachandran Srinivasan
- Centre for Ocean Research, Sathyabama Research Park, Sathyabama Institute of Science and Technology, Chennai 600119, Tamil Nadu, India;
| | - Prathap Somu
- Department of Biotechnology and Chemical Engineering, School of Engineering, Faculty of Science, Technology and Architecture (FoSTA), Manipal University Jaipur, Dehmi Kalan, Off. Jaipur-Ajmer Expressway, Jaipur 303007, Rajasthan, India
| | - Sonaimuthu Mohandoss
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea; (K.S.V.); (M.A.)
- Centre of Molecular Medicine and Diagnostics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| |
Collapse
|
14
|
Yang Y, Tan B, Xiao L, Huang H, Xu W, Su J, Zhao Y, Hong L, Cai G, Li Z, Dai L, Gu T. GPCPD1 is regulated by METTL3/IGF2BP3 dependent m 6A modification to affect pig muscle fiber type conversion. Gene 2025; 962:149578. [PMID: 40404071 DOI: 10.1016/j.gene.2025.149578] [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: 04/18/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025]
Abstract
The N6-methyladenosine (m6A) modification is the most prevalent and abundant RNA modification in eukaryotes. In our previous study, we identified that the glycerophosphocholine phosphodiesterase 1 (GPCPD1) gene was differentially expressed and diverse m6A modificated in the pig soleus and extensor digitorum longus muscles. In this study, we further investigated the function of GPCPD1 gene in pig muscle development. We found that GPCPD1 inhibited myogenic differentiation and promoted the conversion of fast-twitch to slow-twitch fibers in both porcine muscle satellite cells (PSCs) and in mouse model. We also found that the expression of GPCPD1 was affected by m6A methyltransferase METTL3. The methylated GPCPD1 gene was recognized by the m6A reader protein IGF2BP3, which further modulated the stability of GPCPD1 mRNA. This study provided novel evidence into m6A regulation in pig muscle development and illustrated the new post-transcriptional regulation way in GPCPD1 expression.
Collapse
Affiliation(s)
- Yuming Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Baohua Tan
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liyao Xiao
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Huijun Huang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Weihua Xu
- College of Life Science, Longyan University, Longyan, China
| | - Jiawei Su
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Yaolu Zhao
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Linjun Hong
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Gengyuan Cai
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Zicong Li
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Lihe Dai
- Key Laboratory of Animal Genetics and Breeding of Zhejiang Province, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China.
| | - Ting Gu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China.
| |
Collapse
|
15
|
Zheng CH, Chen YB, Yu WB, Cai LS, Wang Q, Sun YH, Yan S, He XL, Xu ZK, Li GX, Tian YT, Li C, Wang BG, Ji JF, Xu YC, Zhong Q, Liu ZY, Chen QY, Li P, Xie JW, Liang Y, Liu ZM, Qiu HB, Wei M, Yan ZB, Lv CB, Chen QX, Li S, Zeng LX, Huo BW, Li ZY, Su XQ, Huang CM. Safety and efficacy of indocyanine green-guided laparoscopic lymphadenectomy for locally advanced gastric cancer: The CLASS-11 clinical trials. Cell Rep Med 2025; 6:102136. [PMID: 40398388 DOI: 10.1016/j.xcrm.2025.102136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/31/2024] [Accepted: 04/22/2025] [Indexed: 05/23/2025]
Abstract
We report the short-term results of indocyanine green (ICG)-guided laparoscopic lymphadenectomy for gastric cancer (GC). The primary outcome is 3-year disease-free survival. In this analysis, we present short-term secondary outcomes focused on the number of lymph nodes (LNs) retrieved and the diagnostic value of fluorescent status for metastatic LNs, excluding long-term outcomes. A total of 1,006 patients are included in the per-protocol analysis. The mean number of LNs retrieved in the ICG group is significantly higher than that in the non-ICG group. The negative predictive value is 93.9% for nonfluorescent stations, and the sensitivity of ICG for detecting all metastatic LN stations is 91.6%. ICG technology is safe and feasible for laparoscopic lymphadenectomy in GC and can noticeably increase the number of LNs retrieved. Further follow-up is necessary to warrant whether ICG can improve long-term survival of GC. The Chinese Laparoscopic Gastrointestinal Surgery Study (CLASS)-11 trial has been registered at ClinicalTrials.gov as NCT04593615.
Collapse
Affiliation(s)
- Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ying-Bo Chen
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wen-Bin Yu
- Department of General Surgery, Qilu Hospital of Shandong University, Shandong, China
| | - Li-Sheng Cai
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Quan Wang
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Jilin, China
| | - Yi-Hong Sun
- Gastric Cancer Center/Department of Gastrointestinal Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Su Yan
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qinghai University, Qinghai, China
| | - Xian-Li He
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ze-Kuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guo-Xin Li
- Department of General Surgery, Southern Medical University Nanfang Hospital, Guangdong, China
| | - Yan-Tao Tian
- Department of Pancreatic and Gastric Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Chen Li
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bao-Gui Wang
- Department of Gastric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Control, Tianjin Clinical Research Center for Cancer, Tianjin, China
| | - Jia-Fu Ji
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yan-Chang Xu
- Gastrointestinal Surgery Unit 1, Teaching Hospital of Putian First Hospital of Fujian Medical University, Fujian, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhi-Yu Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yao Liang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhi-Min Liu
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hai-Bo Qiu
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Meng Wei
- Department of General Surgery, Qilu Hospital of Shandong University, Shandong, China
| | - Zhi-Bo Yan
- Department of General Surgery, Qilu Hospital of Shandong University, Shandong, China
| | - Chen-Bin Lv
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Qiu-Xian Chen
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Shuang Li
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Jilin, China
| | - Ling-Xiao Zeng
- Gastric Cancer Center/Department of Gastrointestinal Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bo-Wen Huo
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qinghai University, Qinghai, China
| | - Zi-Yu Li
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiang-Qian Su
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
16
|
Martviset P, Chantree P, Tongsiri N, Plengsuriyakarn T, Na-Bangchang K. Targeting tumor-associated genes, immune response, and circulating tumor cells in intrahepatic cholangiocarcinoma: Therapeutic potential of Atractylodes lancea (Thunb.) DC. PLoS One 2025; 20:e0323732. [PMID: 40359186 PMCID: PMC12074528 DOI: 10.1371/journal.pone.0323732] [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: 12/06/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Cholangiocarcinoma (CCA) is one of the most aggressive cancers with a poor prognosis. Current treatment strategies involve hepatobiliary surgery, chemotherapy, radiotherapy, and supportive care; however, the success of these treatments remains limited. Therefore, this study investigated the potential of Atractylodes lancea (Thunb) D.C. (AL) in limiting the progress of CCA by targeting the expression of cancer-related genes involved in immune responses and circulating tumor cells. The study was part of Phase 2A clinical trial in advanced-stage intrahepatic iCCA (iCCA) patients: Group 1 (n = 16) received low-dose AL (capsule formulation of the standardized extract of AL: CMC-AL) with standard supportive care, Group 2 (n = 16) received high-dose AL with standard supportive care, and Group 3 (n = 16) received standard supportive care alone. Venous whole blood samples (EDTA, 5 ml) were collected from each patient on Day 1 and Day 90 and the non-CCA subjects (n = 16) on Day 1. Fifty-nine samples (48 and 11 samples for Day 1 and Day 90, respectively) were processed for total RNA isolation. Gene expression was evaluated using reverse transcription followed by a PCR array. Regardless of dosage, gene expression patterns in the AL-treated groups closely resembled those of the healthy subjects. Specifically, cancer-associated genes, including VEGF-A, NR4A3, Ki-67, and EpCAM, were significantly down-regulated. Additionally, the expression levels of immune-related genes were modulated in AL-treated patients. The treatment groups exhibited lower levels of the pro-inflammatory cytokine IL-6, increased expression of the anti-inflammatory cytokine IL-10, and cell-mediated immune-related molecules such as CTLA4 and PFR1. These findings suggest the potential of AL for iCCA treatment. However, additional studies are required to confirm the correlation between gene and protein expression profiles, as well as CTCs profile.
Collapse
Affiliation(s)
- Pongsakorn Martviset
- Division of Parasitology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
- Graduate Program in Applied Biosciences, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Pathanin Chantree
- Graduate Program in Applied Biosciences, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
- Division of Anatomy, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Nisit Tongsiri
- Department of Surgery, Sakon Nakhon Hospital, Sakon Nakhon, Thailand
| | - Tullayakorn Plengsuriyakarn
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
- Center of Excellence in Malaria and Cholangiocarcinoma, Thammasat University, Pathum Thani, Thailand
| | - Kesara Na-Bangchang
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
- Center of Excellence in Malaria and Cholangiocarcinoma, Thammasat University, Pathum Thani, Thailand
| |
Collapse
|
17
|
Chen Q, Wu Z, Ma Y. Transcriptomics-based exploration of ubiquitination-related biomarkers and potential molecular mechanisms in laryngeal squamous cell carcinoma. BMC Med Genomics 2025; 18:84. [PMID: 40355841 PMCID: PMC12070575 DOI: 10.1186/s12920-025-02148-x] [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: 03/19/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND One of the most common and prevalent cancers is laryngeal squamous cell carcinoma (LSCC), which poses a great threat to the life and health of the patient. Nonetheless, it has been demonstrated that ubiquitination is crucial for the development and course of LSCC. Therefore, it is particularly important to identify biomarkers for ubiquitination-related genes (UbRGs) in LSCC. METHODS Differentially expressed genes (DEGs) in the LSCC versus controls were obtained by differential expression analysis. Also, key modular genes associated with LSCC were obtained using weighted gene co-expression network analysis (WGCNA). Next, DEGs, key module genes, and UbRGs were taken to intersect to obtain candidate genes. And then machine algorithms were to screen potential biomarkers, further their diagnostic value were analyzed and validated. Then, therapeutic agents for biomarkers were predict. In addition, the regulatory networks of the biomarkers were mapped. The expression levels of biomarkers were detected in clinical samples using reverse transcription-quantitative PCR (RT-qPCR). RESULTS A total of eight candidate genes were acquired by the overlap 1,911 DEGs, the key modular genes of WGCNA, and 1,393 UbRGs. A sum of four biomarkers (WDR54, KAT2B, NBEAL2 and LNX1) were identified by two machine learning, then these four biomarkers were validated in GSE127165 and the expression trend was consistent with TCGA-LSCC, they were recorded as biomarkers. Moreover, the accuracy of the biomarkers in predicting clinical aspects of LSCC was confirmed by the receiver operating characteristic (ROC) curves. Subsequently, cancers such as malignant neoplasms, colorectal cancers, tumors, and primary malignant neoplasms were significantly associated with the biomarkers, which further suggests that these four biomarkers were strongly associated with cancer. Meanwhile, the drugs garcinol, cocaine, and triazolam, among others, used for LSCC treatment were predicted. Finally, transcription factors (TFs) (BRD4, MYC, AR, and CTCF) were predicted to regulate the biomarkers. RT-qPCR assays illustrated that the expression trends of KAT2B, LNX1 and NBEAL2 remained consistent with the dataset. CONCLUSION The identification of four biomarkers (WDR54, KAT2B, NBEAL2 and LNX1) associated with UbRGs could ultimately serve as a predictive clinical diagnosis of LSCC and provide insight into the molecular mechanisms of LSCC.
Collapse
Affiliation(s)
- Qiu Chen
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, 28 Guiyi Street, Yunyan District, Guiyang, Guizhou, 550004, China
| | - Zhimin Wu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yifei Ma
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, 28 Guiyi Street, Yunyan District, Guiyang, Guizhou, 550004, China.
| |
Collapse
|
18
|
Li S, Liu Y, Gao Z, An C, Gu H, Yin H, Fu R, Shi L, Xue W, Fan D, Fei Q. Methane Valorization to Antioxidant Polysaccharides by Methanotrophic Bacteria. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11019-11029. [PMID: 40274597 DOI: 10.1021/acs.jafc.5c01087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Bioconversion of methane into functional polysaccharides presents a promising strategy for mitigating methane-induced greenhouse gas emissions and addressing the limitations of plant-derived polysaccharide and sugar-based microbial polysaccharides production. In this study, the novel methane-derived polysaccharide (MePS) was obtained from a newly isolated methanotrophic bacterium Alkalicoccus glycogenes WONF2802. Structural characterization found that MePS is a branched-chain glucan with a weight-average molecular weight of 283.2 kDa. Additionally, MePS exhibited considerable antioxidant capacities in both in vitro biochemical assays and the H2O2-induced oxidative stress cell model. This work establishes a potential pathway for polysaccharide production, reducing reliance on plant and sugar-based feedstocks, while offering a new strategy for methane emission reduction.
Collapse
Affiliation(s)
- Shen Li
- Xi'an Key Laboratory of C1 Compound Bioconversion Technology, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yuan Liu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China
| | - Zixi Gao
- Xi'an Key Laboratory of C1 Compound Bioconversion Technology, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Chao An
- Shaanxi Institute of Microbiology, Xi'an 710043, China
| | - Hui Gu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Heng Yin
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Rongzhan Fu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China
- Xi'an Giant Biogene Technology Co., Ltd., Xi'an 710077, China
| | - Liang Shi
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China
| | - Wenjiao Xue
- Shaanxi Institute of Microbiology, Xi'an 710043, China
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China
| | - Qiang Fei
- Xi'an Key Laboratory of C1 Compound Bioconversion Technology, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
19
|
Zhu W, Zhang J, Pan S, Zhang R, Zhang Y, Yang Q, Hu C, Xu Z. Identifying beneficial gastric cancer patient on prognosis after treating with perioperative or postoperative-only chemotherapy: a single-center real-world study. World J Surg Oncol 2025; 23:177. [PMID: 40320534 PMCID: PMC12051334 DOI: 10.1186/s12957-025-03827-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/25/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND This study evaluates the impact of perioperative S-1 and oxaliplatin (SOX) versus postoperative SOX or capecitabine and oxaliplatin (XELOX) on patient prognosis to identify suitable candidates for each therapy. METHOD A retrospective real-world cohort study was conducted using data from Zhejiang Cancer Hospital on gastric cancer patients treated between 2010 and 2019. Patients were divided into perioperative SOX and postoperative SOX or XELOX groups. Propensity score matching (PSM) was used to control for selection bias. Overall survival (OS) was the primary outcome, analyzed using the Kaplan-Meier method and Cox regression. RESULT A total of 816 patients were included: 293 in the perioperative SOX group and 523 in the postoperative chemotherapy group (408 SOX and 115 XELOX). In the perioperative SOX group, the tumor regression grade (TRG) 2-3 subgroup demonstrated a significantly worse overall survival (OS) compared to the postoperative XELOX group (95% CI = 1.064-3.444, P = 0.027). Subgroup analysis revealed that older patients (95% CI = 0.210-0.950, P = 0.036), and those at the cT3 (95% CI = 0.05-1.19, P = 0.008) stage experienced greater benefits from postoperative chemotherapy. When comparing the benefited populations, it was found that patients with CA125 positivity had an advantage trend with adjuvant chemotherapy compared to perioperative SOX chemotherapy. CONCLUSION Real-world data suggest that perioperative SOX chemotherapy does not benefit all patients with advanced gastric cancer. Patients with TRG 2-3, older age, or cT3 stage may achieve better outcomes with postoperative chemotherapy. Additionally, an exploratory analysis indicated that CA125 positivity may be associated with improved survival following adjuvant treatment.
Collapse
Affiliation(s)
- Weiwei Zhu
- Second clinical medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Jiaqing Zhang
- Second clinical medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Siwei Pan
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Ruolan Zhang
- Second clinical medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yanqiang Zhang
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Qing Yang
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Can Hu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China.
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China.
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| | - Zhiyuan Xu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China.
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China.
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| |
Collapse
|
20
|
Yarahmadi HB, Shahryari K, Bozorgi M, Shirdel A, Mohamadi Z, Rooshenas N, Karim Nezhad H, Mobaraki H, Aryannejad M, Emdadi A, Khosravian Y, Shahidi Marnani SA, SadatRafiei SK, Asadi Anar M, Marashi A, Khosravi F, Khodaei M. Dietary inflammatory index and the risk of esophageal cancer: a systematic review and meta-analysis. BMC Cancer 2025; 25:826. [PMID: 40319274 PMCID: PMC12048919 DOI: 10.1186/s12885-025-14199-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND AND AIM It is well-recognized that inflammation is an adaptive pathophysiological response in many types of cancer. Research on nutrition's critical role in inflammation, a risk factor for all forms of cancer, is growing. The dietary inflammatory index (DII) was created lately to assess if a diet is pro- or anti-inflammatory in terms of inflammation. Indeed, several studies have demonstrated the correlation between DII and the risk of several cancer types. This meta-analysis set out to look into the relationship between DII and the different forms of esophageal cancer. METHOD PubMed, Cochrane library, Embase, Scopus, and Web of Science databases were searched up to May 2024 to retrieve relevant articles. RAYYAN intelligent tool for systematic reviews was incorporated for the screening of studies. Original articles written in English Studies that investigated the inflammatory index of diet in individuals who developed esophageal cancer were included in this study.STATA v18 software was used to conduct the meta-analysis. Egger's test for publication bias assessment was implemented. Newcastle Ottawa scale was used to evaluate the qualities of the included studies. A plot digitizer was used to extract digital data. RESULT A total of 13 studies were included in the systematic review, with 6 studies contributing to the meta-analysis, comprising 10,150 participants. The participants were categorized into high and low DII groups, with the low DII group (n = 3,403) serving as the reference. The meta-analysis demonstrated a significant association between high DII and increased risk of esophageal cancer. Specifically, individuals in the high DII group were 29% more likely to develop esophageal cancer, with a pooled odds ratio (OR) of 1.29 (95% Confidence Interval [CI]: 1.16-1.43), as calculated using a random-effects model. Moderate heterogeneity was observed (I2 > 50%). Egger's test indicated evidence of publication bias (p < 0.05). Subgroup and sensitivity analyses confirmed the robustness of this association across populations and study designs. CONCLUSION our study concludes that a higher level of DII is associated with a higher risk of esophageal cancer development. This study suggests that modifying inflammatory properties of dietary patterns can reduce the risk of incidence of esophageal cancer.
Collapse
Affiliation(s)
| | - Kianoush Shahryari
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Bozorgi
- Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran
| | - Ahmadreza Shirdel
- student research committee, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Zhina Mohamadi
- Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Negar Rooshenas
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Helia Karim Nezhad
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Hesam Mobaraki
- Faculty of Medicine, İstanbul Yeniyuzyil University, Istanbul, Turkey
| | - Majid Aryannejad
- Dental School, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Anahita Emdadi
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Yekta Khosravian
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | | | | | - Mahsa Asadi Anar
- College of Medicine, University of Arizona, 1501 N Campbell Ave, Tucson, AZ, 85724, USA.
| | - Amir Marashi
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farbod Khosravi
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Khodaei
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Chen H, Hou S, Zhang H, Zhou B, Xi H, Li X, Lufeng Z, Guo Q. RETRACTED: MiR-375 impairs breast cancer cell stemness by targeting the KLF5/G6PD signaling axis. ENVIRONMENTAL TOXICOLOGY 2025; 40:E31-E43. [PMID: 38470012 DOI: 10.1002/tox.24204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 03/13/2024]
Abstract
Recurrence of breast cancer may be due to the presence of breast cancer stem cells (BCSC). Abnormal tumor cell growth is closely associated with increased reactive oxygen species (ROS) and disruption of redox homeostasis, and BCSCs exhibit low levels of ROS. The detailed mechanism between the low levels of ROS in BCSCs and their maintenance of stemness characteristics has not been reported. A growing number of studies have shown that tumor development is often accompanied by metabolic reprogramming, which is an important hallmark of tumor cells. As the first rate-limiting enzyme of pentose phosphate pathway (PPP), the expression of G6PD is precisely regulated in tumor cells, and there is a certain correlation between PPP and BCSCs. MiR-375 has been shown to inhibit stem cell-like properties in breast cancer, but the exact mechanism is not clear. Here, KLF5, as a transcription factor, was identified to bind to the promoter of G6PD to promote its expression, whereas miR-375 inhibited the expression of KLF5 by binding to the 3'UTR region of KLF5 mRNA and thus reduced the expression of G6PD expression, inhibits PPP to reduce NADPH, and increases ROS levels in breast cancer cells, thereby weakening breast cancer cell stemness. Our study reveals the specific mechanism by which miR-375 targets the KLF5/G6PD signaling axis to diminish the stemness of breast cancer cells, providing a therapeutic strategy against BCSCs.
Collapse
Affiliation(s)
- Haitao Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shanshan Hou
- Department of Pharmacy, Zhejiang Pharmaceutical University, Ningbo, People's Republic of China
| | - Hongwei Zhang
- Department of Anesthesiology, Hepatobiliary Surgery, Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Bing Zhou
- Department of Anesthesiology, Hepatobiliary Surgery, Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Huifang Xi
- Department of Anesthesiology, Hepatobiliary Surgery, Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Xiaofang Li
- Department of Anesthesiology, Hepatobiliary Surgery, Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Zheng Lufeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qianqian Guo
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, People's Republic of China
| |
Collapse
|
22
|
Weng CM, Zhong Q, Sun YQ, Liu ZY, Ma YB, Zhang ZQ, Zhang HX, Zhu JY, Ye W, Wu J, Du H, Zheng CH, Li P, Chen QY, Huang CM, Xie JW. A novel ypN-TRG staging system for gastric cancer patients after neoadjuvant therapy based on the metro-ticket paradigm: a multicenter and large sample retrospective analysis. Gastric Cancer 2025; 28:465-477. [PMID: 39918688 DOI: 10.1007/s10120-025-01586-x] [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: 10/14/2024] [Accepted: 01/15/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Conventional ypT category evaluates the depth of invasion after neoadjuvant therapy (NAT) for gastric cancer (GC) and has limited prognostic value. Tumor regression grade (TRG) measures the extent of tumor response to treatment, and when combined with the ypN category, it may enhance the prediction of patient outcomes. This study aims to develop a new staging system by integrating TRG and ypN category to better evaluate the prognosis of GC patients receiving NAT. METHODS This retrospective analysis included 962 patients who underwent radical gastrectomy after NAT, with 513 in the development cohort (from one center) and 449 in the external validation cohort (from five centers). The ypN-TRG staging system was established by calculating the distance from the origin on a cartesian plane incorporating the ypN (x-axis) stage and TRG (y-axis) grade, and five sub-stages were delineated. RESULTS In the development cohort, 3-year overall survival rates according to ypN-TRG stage I, IIA, IIB, IIIA, IIB were 87.6%, 80.2%, 70.7%, 47.3%, 21.5%, p < 0.01. Compared with ypTNM, the ypN-TRG staging system performed better in terms of the prognostic discrimination power (C-index), goodness-of-fit (AIC, BIC), model improvement (NRI, IDI), and model stability (time-AUC). Multivariate Cox regression analysis confirmed the superiority of ypN-TRG over ypTNM staging. In the external validation cohort, ypN-TRG staging was a better predictor of OS and DFS in patients with GC. CONCLUSIONS The ypN-TRG staging system is superior to the AJCC eighth edition ypTNM staging system in accurately assessing the prognosis of patients with GC after NAT.
Collapse
Affiliation(s)
- Cai-Ming Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Yu-Qin Sun
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, Fujian, China
| | - Zhi-Yu Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qinghai University, Xining, 810000, Qinghai, China
| | - Zhi-Quan Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Hao-Xiang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Ji-Yun Zhu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Wen Ye
- Department of Gastrointestinal Surgery, Longyan First Hospital Affiliated to, Fujian Medical University, Longyan, 364000, Fujian, China
| | - Ju Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116000, Liaoning, China
| | - He Du
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qinghai University, Xining, 810000, Qinghai, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
23
|
Gostomczyk K, Drozd M, Marsool Marsool MD, Pandey A, Tugas K, Chacon J, Tayyab H, Ullah A, Borowczak J, Szylberg Ł. Biomarkers for the detection of circulating tumor cells. Exp Cell Res 2025; 448:114555. [PMID: 40228709 DOI: 10.1016/j.yexcr.2025.114555] [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: 10/22/2024] [Revised: 04/05/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Circulating tumor cells (CTCs) have emerged as a key biomarker in cancer detection and prognosis, and their molecular profiling is gaining importance in precision oncology. Liquid biopsies, which allow the extraction of CTCs, circulating tumor DNA (ctDNA) or cell-free DNA (cfDNA), have measurable advantages over traditional tissue biopsies, especially when molecular material is difficult to obtain. However, this method is not without limitations. Difficulties in differentiating between primary and metastatic lesions, uncertain predictive values and the complexity of the biomarkers used can prove challenging. Recently, high cell heterogeneity has been identified as the main obstacle to achieving high diagnostic accuracy. Because not all cells undergo epithelial-mesenchymal transition (EMT) at the same time, there is a large population of hybrid CTCs that express both epithelial and mesenchymal markers. Since traditional diagnostic tools primarily detect epithelial markers, they are often unable to detect cells with a hybrid phenotype; therefore, additional markers may be required to avoid false negatives. In this review, we summarize recent reports on emerging CTCs markers, with particular emphasis on their use in cancer diagnosis. Most of them, including vimentin, TWIST1, SNAI1, ZEB1, cadherins, CD44, TGM2, PD-L1 and GATA, hold promise for the detection of CTCs, but are also implicated in cancer progression, metastasis, and therapeutic resistance. Therefore, understanding the nature and drivers of epithelial-mesenchymal plasticity (EMP) is critical to advancing our knowledge in this field.
Collapse
Affiliation(s)
- Karol Gostomczyk
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, Poland; Department of Tumor Pathology and Pathomorphology, Oncology Center - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland; Department of Pathology, Dr Jan Biziel Memorial University Hospital, Bydgoszcz, Poland.
| | - Magdalena Drozd
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, Poland; Department of Pathology, Dr Jan Biziel Memorial University Hospital, Bydgoszcz, Poland
| | | | - Anju Pandey
- Memorial Sloan Kettering Cancer Center, New York, USA
| | | | - Jose Chacon
- American University of Integrative Sciences, Saint Martin, Cole Bay, Barbados
| | | | - Ashraf Ullah
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jędrzej Borowczak
- Department of Clinical Oncology, Oncology Center - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland
| | - Łukasz Szylberg
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, Poland; Department of Tumor Pathology and Pathomorphology, Oncology Center - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland; Department of Pathology, Dr Jan Biziel Memorial University Hospital, Bydgoszcz, Poland
| |
Collapse
|
24
|
Sanghvi G, R R, Kashyap A, Sabarivani A, Ray S, Bhakuni PN. Identifying the function of kinesin superfamily proteins in gastric cancer: Implications for signal transduction, clinical significance, and potential therapeutic approaches. Clin Res Hepatol Gastroenterol 2025; 49:102571. [PMID: 40064398 DOI: 10.1016/j.clinre.2025.102571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025]
Abstract
Gastric cancer (GC), a leading cause of cancer-related mortality, poses a significant global health challenge. Given its complex etiology, understanding the molecular pathways driving GC progression is crucial for developing innovative therapeutic strategies. Among the diverse proteins involved in cellular transport and mitotic regulation, kinesin superfamily proteins (KIFs) have emerged as key players in tumor biology. These motor proteins mediate intracellular transport along microtubules and are essential for processes such as cell division, signaling, and organelle distribution. Evidence indicates that specific KIFs are dysregulated in GC, potentially driving cancer cell proliferation, metastasis, and chemoresistance. Moreover, aberrant KIF expression has been associated with poorer prognoses, highlighting their potential as biomarkers for early diagnosis and therapeutic intervention. This review explores the roles of KIFs in GC and assesses their implications for research and clinical applications. By elucidating the significance of KIFs in GC, this discussion aims to inspire novel insights in cancer biology and advance targeted therapeutic strategies.
Collapse
Affiliation(s)
- Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Aditya Kashyap
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India
| | - A Sabarivani
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751003, India
| | - Pushpa Negi Bhakuni
- Department of Allied Science, Graphic Era Hill University, Bhimtal, Uttarakhand 248002, India; Graphic Era Deemed to be University, Dehradun, Uttarakhand, India.
| |
Collapse
|
25
|
Li P, Dong J, Li Y, Yan J, Wang J, Cao S, Cao W, Zhao X, Xue A, Xu Z, Yang L. The impact of ADAMTS14 genetic polymorphisms and its function on susceptibility to and prognosis of gastric cancer in a Chinese Han population. Gastric Cancer 2025; 28:326-343. [PMID: 39992602 DOI: 10.1007/s10120-025-01598-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/03/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND Single nucleotide polymorphisms (SNPs) are associated with various diseases, including gastric cancer. The ADAMTS14 gene is linked to multiple types of cancer. However, the relationship between ADAMTS14 and its genetic polymorphisms with susceptibility to gastric cancer (GC) and prognosis remains unclear. METHODS A case-control study was conducted involving 855 patients diagnosed with gastric cancer (GC) and an equal number of cancer-free controls. Following rigorous statistical analysis, molecular experiments were performed to elucidate the functional significance of the SNPs in the context of GC. RESULTS ADAMTS14 rs3740440 (OR = 1.45, p = 0.014) shows a significant association with increased GC risk, while rs11572 (OR = 0.42, p < 0.001) is associated with protection against GC. Moreover, patients with the (CG + GG) genotype of rs3740440 exhibit a poor prognosis (HR = 1.68, p = 0.007). Mechanistically, luciferase reporter assays revealed that the G allele of rs3740440 disrupts the binding of hsa-miR-4294 and hsa-miR-3198 to the 3' untranslated region (3' UTR) of ADAMTS14, leading to increased expression of ADAMTS14 and the promotion of malignant behaviors in GC cells. CONCLUSIONS Our findings underscore the significant role of ADAMTS14 SNPs in both the risk and prognosis of gastric cancer (GC), providing valuable insights into the underlying molecular mechanisms. Specifically, rs3740440 disrupts the interaction between ADAMTS14 and miRNA, resulting in increased expression of ADAMTS14. This heightened expression enhances its malignant biologic behaviors, indicating that rs3740440 could be a potential predictive marker for gastric cancer risk and prognosis.
Collapse
Affiliation(s)
- Pengyu Li
- Department of General Surgery, the First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Jiacheng Dong
- Department of General Surgery, the First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Yuan Li
- Department of General Surgery, the First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Jiang Yan
- Department of General Surgery, Liyang People's Hospital, Liyang Branch Hospital of Jiangsu Province Hospital, Liyang, 213300, Jiangsu Province, China
| | - Jiawei Wang
- Department of General Surgery, the First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Shuqing Cao
- Department of General Surgery, the First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Wei Cao
- Department of General Surgery, the First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Xinyu Zhao
- Department of General Surgery, the First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Ao Xue
- Department of General Surgery, the First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Zekuan Xu
- Department of General Surgery, the First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Li Yang
- Department of General Surgery, the First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
26
|
Saleh RO, Aboqader Al-Aouadi RF, Almuzaini NA, Uthirapathy S, Sanghvi G, Soothwal P, Arya R, Bareja L, Mohamed Abdelgawwad El-Sehrawy AA, Hulail HM. Glucose metabolism is controlled by non-coding RNAs in autoimmune diseases; a glimpse into immune system dysregulation. Hum Immunol 2025; 86:111269. [PMID: 39999745 DOI: 10.1016/j.humimm.2025.111269] [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: 10/10/2024] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
The immune system accidentally targets the body's tissues, causing inflammation and tissue damage, the root causes of autoimmune illnesses. In recent studies, non-coding RNAs have been shown to significantly control gene expression and metabolic pathways linked to autoimmune diseases. This review investigates the effects of non-coding RNA on glucose metabolism, a route frequently dysregulated in autoimmune illnesses such as multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, and diabetes. We review how non-coding RNA affects immune cell activity modulation, glucose absorption, glycolysis, and other metabolic processes critical to immune function. We also investigate the possibility of using non-coding RNA-mediated metabolic pathway targeting as a new therapeutic approach to treat autoimmune disorders. By clarifying the complex interplay of non-coding RNA, glucose metabolism, and immune dysregulation, this study endeavors to enhance comprehension of autoimmune etiology and facilitate the creation of focused therapies.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al Maarif, Anbar, Iraq
| | | | | | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Pradeep Soothwal
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Renu Arya
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Lakshay Bareja
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India
| | | | - Hanen Mahmod Hulail
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
27
|
Aakel N, Mohammed R, Fathima A, Kerzabi R, Abdallah A, Ibrahim WN. Role of Exosome in Solid Cancer Progression and Its Potential Therapeutics in Cancer Treatment. Cancer Med 2025; 14:e70941. [PMID: 40344389 PMCID: PMC12063069 DOI: 10.1002/cam4.70941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/14/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Exosomes are extracellular vesicles ranging from 40 to 100 nm in diameter that mediate intercellular communication by transferring proteins, lipids, nucleic acids, and other metabolites. In the context of cancer, exosomes influence the tumor microenvironment by carrying regulatory RNAs such as miRNA, circRNA, and lncRNA. They originate from various cells, including adipocytes, fibroblasts, and hepatocellular carcinoma (HCC) cells, and can either promote or inhibit cancer progression through pathways like MAPK and PI3K-Akt. AIM This review aims to explore the role of exosomes in the progression of solid cancers, emphasizing their self-induced activation mechanisms and how they modulate tumor behavior. METHODOLOGY A comprehensive review of recent literature was conducted, focusing on studies that investigated the biological functions of exosomes in solid tumor progression, including their molecular cargo, cellular origin, and involvement in signaling pathways. RESULTS Findings from multiple studies indicate that cancer-derived exosomes contribute to tumor proliferation, metastasis, and therapy resistance by enhancing communication within the tumor microenvironment. These vesicles activate oncogenic pathways and can serve as biomarkers or therapeutic targets due to their role in disease modulation. CONCLUSION Exosomes play a pivotal role in solid cancer progression and offer significant potential in advancing our understanding of tumor biology. Their capacity to influence key signaling pathways and facilitate intercellular communication makes them promising candidates for novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Nada Aakel
- Department of Biomedical ScienceCollege of Health Sciences, QU Health, Qatar UniversityDohaQatar
| | - Rawdhah Mohammed
- Department of Biomedical ScienceCollege of Health Sciences, QU Health, Qatar UniversityDohaQatar
| | - Assela Fathima
- Department of Biomedical ScienceCollege of Health Sciences, QU Health, Qatar UniversityDohaQatar
| | - Rabia Kerzabi
- Department of Biomedical ScienceCollege of Health Sciences, QU Health, Qatar UniversityDohaQatar
| | - Atiyeh Abdallah
- Department of Biomedical ScienceCollege of Health Sciences, QU Health, Qatar UniversityDohaQatar
| | - Wisam Nabeel Ibrahim
- Department of Biomedical ScienceCollege of Health Sciences, QU Health, Qatar UniversityDohaQatar
| |
Collapse
|
28
|
Wu Z, Huang B, Zhang D, Yu Q, Yan C. Structural characterization of a pectin-like polysaccharide from Clematis chinensis Osbeck and its anti-rheumatoid arthritis activity. Int J Biol Macromol 2025; 307:141917. [PMID: 40068746 DOI: 10.1016/j.ijbiomac.2025.141917] [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: 10/27/2024] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/25/2025]
Abstract
Clematis chinensis Osbeck is a traditional Chinese medicine to treat rheumatic arthritis (RA). We hypothesized that C. chinensis polysaccharide, as a bioactive ingredient, might have the anti-RA function. However, the structure and bioactivity of C. chinensis polysaccharides are less explored. A pectin-like polysaccharide CCPB-2-2 was isolated from C. chinensis root. The putative chemical structure of CCPB-2-2 was characterized through monosaccharide composition, uronic acid reduction, partial acid hydrolysis, methylation reaction, and nuclear magnetic resonance spectroscopy. CCPB-2-2 was composed of the RG-I domain of the pectin moiety and the non-pectin moiety with a backbone with →3)-β-D-Galp-(1→, →6)-β-D-Galp-(1→, →4)-α-D-GlcAp-(1→, →4)-α-D-Glcp-(1→, →4,6)-α-D-Glcp-(1→, →4)-β-D-Galp-(1→, and →3,6)-α-D-Manp-(1→ residues. Moreover, the anti-RA activity of CCPB-2-2 was evaluated in MH7A cells. CCPB-2-2 could inhibit lipopolysaccharide-induced MH7A cell proliferation, down-regulate the expression of pro-inflammatory factors (interleukin-6, interleukin-8, and interleukin-1 beta) and matrix metalloproteinase genes (matrix metallopeptidase 1 and matrix metallopeptidase 3), regulate apoptosis proteins (B-cell lymphoma-2-associated X and B-cell lymphoma-2) and inhibit cell migration. In conclusion, C. chinensis polysaccharide CCPB-2-2 exhibited great potential as a drug candidate for the treatment of RA, which is worth of further research and development.
Collapse
Affiliation(s)
- Zhijian Wu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Binhua Huang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dawei Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Qian Yu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Chunyan Yan
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
29
|
Li Q, Zhang R, Ouyang C, Wang S, Li S, Yin X, Deng Z, Han B, Chi J. Photocurable Dual-Network Hydrogels Based on Natural Polymers for Sutureless Repair of Large Corneal Defects. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2500150. [PMID: 40159843 DOI: 10.1002/smll.202500150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Corneal transplantation remains the prevailing treatment for corneal defects, which is always restricted by donor shortages and numerous postoperative complications accompanying suturing. Photocurable hydrogels have emerged as alternative therapeutic strategies for the repair of corneal defects, but most hydrogels focus on repairing focal corneal defects and still suffer from low transparency and poor mechanical properties. Herein, photocurable hydrogel GelMA/OCS composed of gelatin methacryloyl (GelMA) and oxidized chondroitin sulfate (OCS) is developed for sutureless repair of large corneal defects (6 mm). This injectable hybridized hydrogel demonstrates excellent transparency, low swelling rate, enhanced mechanical properties, and superior adhesion properties. In vitro experiments reveal that GelMA/OCS hydrogel can support the proliferation and migration, and adhesion growth of human corneal epithelial cells (HCECs), demonstrating satisfactory cytocompatibility and cell affinity. In addition, GelMA/OCS hydrogel is capable of accurately filling the large corneal defects in rabbits and forming hydrogel grafts with smooth surfaces. Postoperative slit lamp, histological evaluation, and transcriptomic analysis reveal that GelMA/OCS hydrogel can significantly facilitate corneal re-epithelialization and the integration and reconstruction of stromal structures, as well as reduce inflammation responses and scar formation. Therefore, GelMA/OCS hydrogel may provide a promising alternative for the sutureless treatment of large corneal defects.
Collapse
Affiliation(s)
- Qing Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Ruyin Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Chengpei Ouyang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Shuo Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Shanshan Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Xinchao Yin
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Zimeng Deng
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Baoqin Han
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Jinhua Chi
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| |
Collapse
|
30
|
Liu S, Wan X, Gou Y, Yang W, Xu W, Du Y, Peng X, Wang X, Zhang X. The emerging functions and clinical implications of circRNAs in acute myeloid leukaemia. Cancer Cell Int 2025; 25:167. [PMID: 40296024 PMCID: PMC12038945 DOI: 10.1186/s12935-025-03772-4] [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: 04/28/2024] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Acute myeloid leukaemia (AML) is a prevalent haematologic malignancy characterized by significant heterogeneity. Despite the application of aggressive therapeutic approaches, AML remains associated with poor prognosis. Circular RNAs (circRNAs) constitute a unique class of single-stranded RNAs featuring covalently closed loop structures that are ubiquitous across species. These molecules perform crucial regulatory functions in the pathogenesis of various diseases through diverse mechanisms, including acting as miRNA sponges, interacting with DNA or proteins, and encoding functional proteins/polypeptides. Recently, numerous circRNAs have been confirmed to have aberrant expression patterns in AML patients. In particular, certain circRNAs are closely associated with specific clinicopathological characteristics and thus have great potential as diagnostic/prognostic biomarkers and therapeutic targets in AML. Herein, we systematically summarize the biogenesis, degradation, and functional mechanisms of circRNAs while highlighting their clinical relevance. We also outline a series of online databases and analytical tools available to facilitate circRNA research. Finally, we discuss the current challenges and future research priorities in this evolving field.
Collapse
Affiliation(s)
- Shuiqing Liu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Xingyu Wan
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Yang Gou
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Wuchen Yang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Wei Xu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Yuxuan Du
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Xiangui Peng
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400037, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
31
|
Kim EM, Hong ST. Clonorchis sinensis and Cholangiocarcinoma. J Korean Med Sci 2025; 40:e145. [PMID: 40296827 PMCID: PMC12040606 DOI: 10.3346/jkms.2025.40.e145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Clonorchiasis is a parasitic disease caused by Clonorchis sinensis, a trematode that inhabits the intrahepatic bile ducts of humans and mammals. C. sinensis is one of common food-borne trematodes, prevalent in East Asia including Korea. The International Agency for Research on Cancer reclassified C. sinensis as the Group 1 biological carcinogen of human cholangiocarcinoma (CCA). Evidence supporting the carcinogenicity of C. sinensis includes epidemiological studies showing increased prevalence and odds ratio (OR) of CCA in clonorchiasis patients, the development of CCA in experimental animals, and molecular studies. Approximately 10% of CCA in Korea are believed to be solely caused by clonorchiasis, with an OR of 4.7 for CCA risk among clonorchiasis patients. All hamsters exposed to both of C. sinensis and N-nitrosodimethylamine (NDMA) developed CCA while those exposed to either C. sinensis or NDMA alone did not. In vitro studies using cell models investigated carcinogenetic changes of the intracellular molecules and genes following stimulation with a soluble extract of C. sinensis. The in vitro stimulated cells showed a significant shift to G2/M phage, produced oncogenic molecules, changed expression of oncogenes, increased cell proliferation and suppressed apoptosis. Additionally, the gap-junction proteins between cells, such as connexin (Cx) 43, Cx 26, and Cx 32, were changed significantly, disrupting intercellular homeostasis. These findings suggest that C. sinensis and nitrogen compounds synergistically stimulate the cholangiocytes to become neoplastic. C. sinensis is a biological carcinogen of human CCA, and the World Health Organization guidelines enlist food-borne trematodes as one of target neglected tropical diseases to be eliminated by 2030. The present article reviews and updates perspectives on clonorchiasis, focusing on carcinogenesis.
Collapse
Affiliation(s)
- Eun-Min Kim
- Department of Microbiology, College of Medicine and Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Sung-Tae Hong
- Department of Tropical Medicine and Parasitology and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
32
|
Asadi S, Grafenauer S, Burley CV, Fitzgerald C, Humburg P, Parmenter BJ. The effectiveness of dietary intervention in osteoarthritis management: a systematic review and meta-analysis of randomized clinical trials. Eur J Clin Nutr 2025:10.1038/s41430-025-01622-0. [PMID: 40295866 DOI: 10.1038/s41430-025-01622-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 03/06/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025]
Abstract
This study aimed to evaluate the impact of various dietary interventions on managing osteoarthritis (OA), a condition significantly affecting global health due to joint alterations driven by inflammatory mediators. A systematic review and meta-analysis, adhering to PRISMA guidelines, examined Randomized Controlled Trials (RCTs) investigating dietary interventions in OA. Two reviewers independently conducted study selection, data extraction, and quality assessment. Random effects models calculated standardized mean differences (SMD) and mean differences (MD). Risk of bias was evaluated with the Cochrane Risk of Bias tool (RoB2), and heterogeneity was assessed using I² values. Nine RCTs (898 participants) were identified, assessing various diets: reduced energy (n = 4), Mediterranean (n = 2), low-fat (n = 2), anti-inflammatory (n = 1), low-carbohydrate (n = 1), and plant-based (n = 1). Dietary interventions significantly improved pain (SMD: -0.67; 95% CI: [-1.01, -0.34]; p < 0.0001), and physical function (SMD: -0.62; 95% CI: [-0.94, -0.30]; p = 0.0001) and body weight (MD: -3.18; 95% CI: [-3.52, -2.83], p < 0.0001). Subgroup analyses revealed reduced energy diets improved pain (SMD: -0.85; 95% CI: [-1.15, -0.55], p < 0.0001), physical function (SMD: -0.95; 95% CI: [-1.33, -0.58], p < 0.0001) and body weight (MD: -3.13; 95% CI: [-3.77, -2.49], p < 0.0001). The Mediterranean diet did not significantly impact pain (SMD: -0.27; 95% CI: [-1.14, 0.60], P = 0.54), or physical function (SMD = -0.28; 95% CI: [-0.79, 0.24], p = 0.29). This study emphasizes the significant impact of dietary interventions on pain, physical function, and weight management in people with OA, with reduced energy diets showing the most effectiveness. Specific dietary patterns show promise but require further investigation.
Collapse
Affiliation(s)
- Sara Asadi
- UNSW Medicine & Health Lifestyle Clinic, School of Health Sciences, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, 2052, Australia
- Department of Exercise Physiology, School of Health Sciences, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Sara Grafenauer
- Department of Nutrition, Dietetics and Food Innovation, School of Health Sciences, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Claire V Burley
- Department of Exercise Physiology, School of Health Sciences, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, 2052, Australia
- Dementia Centre of Excellence, enAble Institute, Curtin University, Perth, WA, Australia
| | - Caroline Fitzgerald
- UNSW Medicine & Health Lifestyle Clinic, School of Health Sciences, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, 2052, Australia
- Department of Exercise Physiology, School of Health Sciences, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Peter Humburg
- Mark Wainwright Analytical Centre, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Belinda J Parmenter
- UNSW Medicine & Health Lifestyle Clinic, School of Health Sciences, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, 2052, Australia.
- Department of Exercise Physiology, School of Health Sciences, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, 2052, Australia.
- School of Health, University of the Sunshine Coast, Sippy Downs, QLD, Australia.
| |
Collapse
|
33
|
Shang-Guan ZX, Zhong Q, Zhang ZQ, Liu ZY, Sun YQ, Ma YB, Ding FH, Wu SC, Zhu JY, Wang YH, Zhao W, Yu JH, Li BL, Wu J, Ye W, Li P, Xie JW, Zheng CH, Chen QY, Huang CM. Surgical and oncological outcomes of laparoscopic versus open gastrectomy after neoadjuvant chemotherapy in patients with locally advanced gastric cancer: A multicenter analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110060. [PMID: 40398315 DOI: 10.1016/j.ejso.2025.110060] [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: 01/31/2025] [Revised: 03/31/2025] [Accepted: 04/13/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Evidence based on large-scale samples comparing the efficacy of laparoscopic gastrectomy (LG) and open gastrectomy (OG) in patients with locally advanced gastric cancer (LAGC) after neoadjuvant chemotherapy (NACT) remains limited. This multicenter study aimed to evaluate the short -term and oncological outcomes of LG and OG after NACT. METHODS Data from a multicenter database of LAGC patients undergoing radical gastrectomy after NACT across 12 centers in China. Propensity score matching (PSM 3:1) was used to balance baseline characteristics. Short-term outcomes, 5-year overall survival (OS), disease-free survival (DFS), and recurrence patterns were compared. RESULTS In total, 962 patients fulfilled the inclusion criteria, of whom 753 underwent LG and 209 underwent OG. After PSM, 627 LG and 209 OG patients were analyzed. Both groups had comparable clinical and pathological characteristics (SMD≤0.100). Intraoperative blood loss was lower in the LG group, with earlier ambulation and diet initiation than that in the OG group (P < 0.05). The number of lymph nodes retrieved was higher in LG group (P < 0.001). Although no significant differences were shown in 5-year OS (LG vs. OG: 51.7vs.43.4 %) and 5-year DFS (LG vs. OG, 48.8 vs. 41.4 %; P > 0.05), landmark analysis revealed improved OS (77.6vs. 65.9 %; P = 0.024) and DFS (84.1vs.71.4 %; P = 0.031) after a landmark time of 28 months in the LG group. Most recurrences occurred within 3 years postoperatively, with similar recurrence patterns in both groups (P > 0.05). CONCLUSIONS In LAGC patients after NACT, LG yielded faster recovery while demonstrating comparable surgical efficacy and oncological outcomes to OG, with potential long-term survival benefits.
Collapse
Affiliation(s)
- Zhi-Xin Shang-Guan
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhi-Quan Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhi-Yu Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yu-Qin Sun
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, China
| | - Fang-Hui Ding
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Shi-Chao Wu
- Gastrointestinal Surgery Unit 2, Teaching Hospital of Putian First Hospital of Fujian Medical University, Putian, China
| | - Ji-Yun Zhu
- Department of Hepato-pancreato-biliary Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yong-Hong Wang
- Department of Gastrointestinal Surgery, The People's Hospital of Leshan, Leshan, China
| | - Wei Zhao
- Department of General Surgery, The General Hospital of Ningxia Medical University, Ningxia, China
| | - Jun-Hua Yu
- Department of General Surgery, The Quzhou Affiliated Hospital of Wenzhou University, Quzhou, China
| | - Bao-Long Li
- Department of General Surgery, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Ju Wu
- Department of General Surgery, The Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Wen Ye
- Department of Gastrointestinal Surgery, The Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
34
|
Guo H, Lou Y, Hou X, Guan X, Guo Y, Han Q, Xue X, Wang Y, He L, Li Z, Zhang C. The mechanistic study of codonopsis pilosula on laryngeal squamous cell carcinoma based on network pharmacology and experimental validation. Front Pharmacol 2025; 16:1542116. [PMID: 40351428 PMCID: PMC12061682 DOI: 10.3389/fphar.2025.1542116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Introduction Laryngeal squamous cell carcinoma (LSCC) is a common malignant tumor of the head and neck, with poor prognosis for advanced patients, and there is an urgent need to find new treatment strategies. Codonopsis pilosula, a traditional Chinese medicinal herb, possesses various pharmacological activities, but its antitumor effects and mechanisms in LSCC are still unclear. The aim of this study was to systematically investigate the potential antitumor mechanism of Codonopsis pilosula in LSCC. Methods In this study, we screened the effective compounds and targets of Codonopsis pilosula by TCMSP, ETCM and BATMAN-TCM databases, and screened targets related to LSCC by combining DisGeNET, GeneCards database and Cytoscape software. KEGG pathway enrichment analysis was utilized to explore the related signaling pathways. The core targets were further screened based on TCGA and GEO database analysis, and molecular docking was carried out to predict their binding ability to effective compounds. The presence of key compounds was verified by LC-MS, the MAPK3 expression was detected by qPCR in LSCC tissues, and the effects of MAPK3 knockdown on proliferation, migration, invasion, cell cycle, and apoptosis of LSCC cells were evaluated by cellular function assays. Results In this study, 22 targets of Codonopsis pilosula that might regulate LSCC were screened based on network pharmacology. KEGG pathway enrichment analysis showed that Codonopsis pilosula-LSCC targets were mainly involved in HIF-1, TNF, IL-17 and FoxO signaling pathways. Based on TCGA and GEO database analysis, MAPK3 was identified as the core target of Codonopsis pilosula-LSCC. The molecular docking results showed that a variety of effective compounds from Codonopsis pilosula had strong binding abilities to MAPK3, among them, Caprylic Acid, Emodin and Luteolin have been confirmed by LC-MS. QPCR analysis indicated that MAPK3 was highly expressed in LSCC tissues. MAPK3 knockdown significantly inhibits LSCC cell proliferation, migration and invasion. It also suppresses LSCC cell growth by blocking the cell cycle and inducing apoptosis. Conclusion Codonopsis pilosula exerts antitumor effects in LSCC through the regulation of MAPK3 and multiple signaling pathways, providing a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Huina Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yichen Lou
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Xiaofang Hou
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Xiaoya Guan
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yujia Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Qi Han
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xuting Xue
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ying Wang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Long He
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhongxun Li
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunming Zhang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Department of Otolaryngology Head and Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
35
|
Liu G, Liu Q, Jia L, Chai Z, Jing L, Xu F, Fan Y. Exosomal circRNAs: key modulators in breast cancer progression. Cell Death Discov 2025; 11:196. [PMID: 40274787 PMCID: PMC12022065 DOI: 10.1038/s41420-025-02494-w] [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: 01/25/2025] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
Breast cancer (BC) poses significant challenges globally, necessitating a deeper understanding of its complexities. Exosomes are cell-specific secreted extracellular vesicles of interest, characterized by a lipid bilayer structure. Exosomes can carry a variety of bioactive components, including nucleic acids, lipids, amino acids, and small molecules, to mediate intercellular signaling. CircRNAs are a novel class of single-stranded RNA molecules, characterized by a closed-loop structure. CircRNAs mainly exert ceRNA functions to intricately modulate gene expression and signaling pathways in breast cancer, influencing tumor progression and therapeutic responses. The unique packaging of circRNAs within exosomes serves as novel genetic information transmitters, facilitating communication between BC cells and microenvironmental cells, thereby regulating critical aspects of BC progression, immune evasion, and drug resistance. Besides, exosomal circRNAs possess the capabilities of serving as diagnostic and therapeutic biomarkers of BC, due to their stability, specificity, and regulatory roles in tumorigenesis and metastasis. Therefore, this review aims to elucidate the novel roles and mechanisms of exosomal circRNAs in BC progression, as well as their potential for diagnosis and therapeutics. The ongoing investigations of exosomal circRNAs will potentially revolutionize treatment paradigms and improve patient outcomes of BC.
Collapse
Affiliation(s)
- Guozhen Liu
- Department of Spinal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Quan Liu
- Department of Thyroid and Breast Surgery, The First People's Hospital of Xiantao, Affiliated Hospital of Hubei University of Science and Technology, Xiantao, China
| | - Lingmei Jia
- Department of Anesthesiology and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zhi Chai
- Clinical Laboratory Center, Xi'an People's Hospital Xi'an Fourth Hospital, Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Li Jing
- School of Basic Medical Sciences, Ningxia Key Laboratory of Vascular Injury and Repair, Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Fangjing Xu
- Department of Critical Care Medicine, Yinchuan Hospital of Traditional Chinese Medicine, Affiliated to Ningxia Medical University, Yinchuan City, Ningxia Hui Autonomous Region, China.
| | - Yucheng Fan
- Department of Pathology, The First People's Hospital of Shizuishan, Affiliated to Ningxia Medical University, Shizuishan City, Ningxia Hui Autonomous Region, China.
| |
Collapse
|
36
|
You GR, Chang DY, Huang HH, Chen YJ, Chang JT, Cheng AJ. Deciphering Nicotine-Driven Oncogenesis in Head and Neck Cancer: Integrative Transcriptomics and Drug Repurposing Insights. Cancers (Basel) 2025; 17:1430. [PMID: 40361356 PMCID: PMC12070984 DOI: 10.3390/cancers17091430] [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] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/18/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Chronic nicotine exposure drives head and neck cancer (HNC) progression, yet its molecular mechanisms remain underexplored. This study examines nicotine-induced transcriptomic changes and potential therapies via drug repurposing. Methods: HNC cell lines (OECM1, SAS, and CGHNC9) were exposed to an IC30 nicotine dose for three months to model chronic exposure in habitual smokers. Transcriptomic profiling of these sublines was integrated with TCGA-HNSC patient data. Differentially expressed genes (DEGs) underwent functional pathway enrichment analysis. Drug repurposing was conducted using gene-drug correlation analysis across GDSC, CTRP, and PRISM databases. Results: Transcriptomic analysis identified 1223 DEGs in nicotine-exposed HNC cells, and integration with TCGA-HNSC data defined a Nic-HNC gene set of 168 genes: 149 oncogenes and 19 tumor suppressors, with 36 oncogenes overexpressed in heavy smokers. Pathway analysis revealed the upregulation of oncogenic signaling, such as PI3K-AKT, alongside the suppression of immune regulation and metabolic reprogramming. Drug repurposing identified five compounds-AZD1332, JAK-8517, NU7441, BRD-K30748066, and neopeltolide-with the first two exhibiting the strongest inverse correlations with nicotine-induced oncogenes in heavy smokers, highlighting their potential as targeted therapies for tobacco-associated HNC. Conclusions: This study comprehensively characterizes nicotine-driven molecular dysregulation in HNC and proposes AZD1332 and JAK-8517 as promising therapeutic candidates through drug repurposing. These insights advance our understanding of nicotine's oncogenic role and provide a foundation for translational research to develop targeted interventions for tobacco-associated HNC.
Collapse
Affiliation(s)
- Guo-Rung You
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (G.-R.Y.); (D.Y.C.); (H.-H.H.)
| | - Daniel Yu Chang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (G.-R.Y.); (D.Y.C.); (H.-H.H.)
| | - Hung-Han Huang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (G.-R.Y.); (D.Y.C.); (H.-H.H.)
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Yin-Ju Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Radiation Oncology and Proton Therapy Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan;
| | - Joseph T. Chang
- Department of Radiation Oncology and Proton Therapy Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan;
- School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ann-Joy Cheng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (G.-R.Y.); (D.Y.C.); (H.-H.H.)
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Department of Radiation Oncology and Proton Therapy Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan;
| |
Collapse
|
37
|
Xu W, Zhu L, Zhang S, Wang X, Gong D, Fan Y. Advances in the roles and molecular mechanisms of exosomal circular RNAs in regulating the pre-metastatic niche of tumors. Discov Oncol 2025; 16:568. [PMID: 40252161 PMCID: PMC12009264 DOI: 10.1007/s12672-025-02374-w] [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: 10/17/2024] [Accepted: 04/11/2025] [Indexed: 04/21/2025] Open
Abstract
Metastasis remains a major cause of morbidity and mortality in patients with malignant tumors. The pre-metastatic niche is a prerequisite for distant metastasis driven by primary tumors. Circular RNAs (circRNAs), a class of single-stranded closed non-coding RNAs, exhibit high stability, evolutionary conservation, and cell-type specificity. Exosomes, as natural carriers of circRNAs, mediate intercellular communication and contribute to the formation of the pre-metastatic niche; however, the mechanisms by which they do so remain incompletely understood. This review summarizes the biological characteristics and functions of exosomal circRNAs and outlines the molecular pathways through which they shape the tumor pre-metastatic microenvironment, with emphasis on immunosuppression, vascular permeability, extracellular matrix remodeling, and lymphangiogenesis. This is the first review to focus on the functional roles and molecular mechanisms of exosomal circRNAs in pre-metastatic niche regulation, providing a basis for the development of therapeutic strategies targeting metastatic progression.
Collapse
Affiliation(s)
- Wei Xu
- Cancer Institute, Affiliated People's Hospital of Jiangsu University, No 8, Dianli Road, Zhenjiang, 212002, Jiangsu, People's Republic of China
| | - Luyu Zhu
- Department of Gastroenterology, Affiliated Suqian First People's Hospital of Nanjing Medical University, No 120, Suzhi Road, Suqian, 223812, Jiangsu, People's Republic of China
| | - Shiqi Zhang
- Department of Gastroenterology, Affiliated Suqian First People's Hospital of Nanjing Medical University, No 120, Suzhi Road, Suqian, 223812, Jiangsu, People's Republic of China
| | - Xiaoyan Wang
- Department of Gastroenterology, Affiliated Suqian First People's Hospital of Nanjing Medical University, No 120, Suzhi Road, Suqian, 223812, Jiangsu, People's Republic of China.
| | - Dandan Gong
- Cancer Institute, Affiliated People's Hospital of Jiangsu University, No 8, Dianli Road, Zhenjiang, 212002, Jiangsu, People's Republic of China.
| | - Yu Fan
- Cancer Institute, Affiliated People's Hospital of Jiangsu University, No 8, Dianli Road, Zhenjiang, 212002, Jiangsu, People's Republic of China.
| |
Collapse
|
38
|
Wang H, Yan C, Ye H. RNF126 Promotes Ovarian Cancer Progression by Reprogramming Lipid Metabolism Through Degradation of ACAP2. Biochem Genet 2025:10.1007/s10528-025-11107-1. [PMID: 40251363 DOI: 10.1007/s10528-025-11107-1] [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: 12/31/2024] [Accepted: 04/12/2025] [Indexed: 04/20/2025]
Abstract
Ovarian cancer (OC) primarily arises from heterogeneous malignant epithelial tissue in the ovary, fallopian tube, or peritoneum. Ubiquitin ligase Ring-finger protein 126 (RNF126) was aberrantly expressed in OC. However, its molecular mechanism is unknown. This study investigates the role and mechanism of RNF126 in regulating ArfGAP with coiled-coil, ankyrin repeat, and PH domains 2 (ACAP2) during OC progression. RT-qPCR and Western blot (WB) were used to assess the expression of RNF126, ACAP2, and lipid synthesis-related genes in OC tissues and cells. The proliferation and migration abilities of OC cells were detected by CCK-8 and Transwell assays. Nile red staining was used to detect the lipid accumulation. The interaction between RNF126 and ACAP2 in OC cells was detected using co-immunoprecipitation (Co-IP). The stability of the ACAP2 protein was analyzed using the cycloheximide (CHX) assay. The effect of RNF126 on tumor growth and metastasis in vivo was investigated by detecting tumor volume and size as well as the number of lung nodules. The expression of RNF126 was upregulated in OC tissues and cells, and RNF126 silencing significantly inhibited the proliferation, migration, and lipid accumulation of OC cells. Mechanistically, ACAP2 was identified as a ubiquitination substrate of RNF126, and its expression was negatively regulated by RNF126. Furthermore, RNF126 promoted OC progression both in vitro and in vivo by suppressing ACAP2 protein expression. RNF126 promotes ovarian cancer progression by reprogramming lipid metabolism via degrading ACAP2.
Collapse
Affiliation(s)
- Hao Wang
- Department of Gynecology and Obstetrics, The First College of Clinical Medical Science, China Three Gorges University, No. 183, Yiling Avenue, Yichang, 443000, Hubei, China
| | - Chao Yan
- Department of Orthopedics, College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
| | - Hong Ye
- Department of Gynecology and Obstetrics, The First College of Clinical Medical Science, China Three Gorges University, No. 183, Yiling Avenue, Yichang, 443000, Hubei, China.
| |
Collapse
|
39
|
Jia F, Wang B, Ma H, Bai C, Zhang Y. Research progress on extraction, separation, structure, and biological activities of polysaccharides from jujube fruit ( Ziziphus jujuba Mill.): a review. Front Chem 2025; 13:1581947. [PMID: 40308264 PMCID: PMC12041218 DOI: 10.3389/fchem.2025.1581947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Jujube (Ziziphus Jujuba Mill.) is an excellent medicinal and edible plant owing to its high nutritional and health-promoting properties. As an important bioactive component, Z. Jujuba polysaccharides have aroused wide attention due to their various pharmacological activities, including anti-inflammatory, immunomodulatory, anti-oxidant, anti-tumor, anti-viral, regulating gut microbiota, hepatoprotective effects and prebiotic activity, and so on. This review highlights the advancements in the extraction methods, structural characteristics, structural elucidation, and functional analysis of polysaccharides derived from Jujube fruits over the past decade, aiming to provide valuable insights for future development and commercialization of Jujube fruits polysaccharides.
Collapse
Affiliation(s)
- Feilong Jia
- Department of Pharmacy, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Bo Wang
- Department of Pharmacy, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Hui Ma
- Department of Pharmacy, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Changcai Bai
- College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yuanyuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
40
|
Wang L, Yang S, Li J, Fang Y, Guo M, Du X, Song L, Chen S, Zhang X, Qi Z, Zhang K, Lv B, Xia J. YAP-activated NAT10 promotes hepatoblastoma progression by activating the pentose phosphate pathway. Int J Biol Sci 2025; 21:2864-2879. [PMID: 40303290 PMCID: PMC12035897 DOI: 10.7150/ijbs.109552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/30/2025] [Indexed: 05/02/2025] Open
Abstract
Hepatoblastoma (HB) is the most common malignant liver tumor in children, with limited treatment options. The N4-acetylcytidine (ac4C) modification, an important mRNA post-transcriptional modification catalyzed by N-acetyltransferase 10 (NAT10), plays a crucial role in the initiation and progression of tumors. However, its impact on the development and prognosis of HB is largely unknown. This study demonstrates that NAT10 is notably upregulated in HB. NAT10 inhibition suppressed HB proliferation and metastasis in vitro and in vivo. Mechanistically, Yes-associated protein 1 (YAP1) induced NAT10 transcription by binding to its promoter, which stimulates the ac4C modification within the 3' untranslated region (3' UTR) of glucose-6-phosphate dehydrogenase (G6PD) and enhancing its mRNA stability. YAP1/NAT10/G6PD axis resulted in enhanced pentose phosphate pathway (PPP) to promote proliferation and metastasis of HB. Moreover, said NAT10-mediated oncogenic effect could be significantly attenuated by a NAT10 inhibitor (Remodelin) both in vitro experiments and in vivo HB mouse models. Overall, our findings revealed the oncogenic role of NAT10 in regulating HB growth and metastasis, which can be a potential therapeutic target for human HB.
Collapse
Affiliation(s)
- Lingxiao Wang
- Department of General Pediatrics, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Shiguang Yang
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai, China
| | - Jie Li
- National Medical Center & National Clinical Research Center for Interventional Medicine, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Fang
- Department of Liver Surgery, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital,Fudan University, Shanghai, China
| | - Mengzhou Guo
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaojing Du
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Li Song
- National Medical Center & National Clinical Research Center for Interventional Medicine, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sinuo Chen
- National Medical Center & National Clinical Research Center for Interventional Medicine, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xingxing Zhang
- Department of Gastroenterology, Shanghai Jiaotong University Affiliated Sixth People Hospital South Campus, Shanghai, China
| | - Zhuoran Qi
- National Medical Center & National Clinical Research Center for Interventional Medicine, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kaihui Zhang
- Institute of Pediatric Research, Children's Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Bei Lv
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinglin Xia
- National Medical Center & National Clinical Research Center for Interventional Medicine, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Ouyang J, Chai H, Sun C, Wang S, She C, Geng D, Xu W. Titanium Particles Activate Osteocytic Connexin 43 to Induce Oxidative Stress and Osteoclastogenesis Through the JAK-STAT Pathway. Antioxid Redox Signal 2025. [PMID: 40207369 DOI: 10.1089/ars.2024.0894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Aims: Periprosthetic osteolysis (PPO), a leading cause of aseptic loosening in joint replacement, arose from complex interactions among osteoblasts, osteoclasts, and osteocytes. Given the pivotal role of connexin 43 (Cx43) in osteocyte communication and bone remodeling, investigating its function was essential for understanding the mechanisms of osteolysis. Our previous studies showed that titanium (Ti) particles increased Cx43 expression in osteocytes. However, the role of Cx43 in osteolysis remained unclear. This study investigated the role of Cx43-mediated regulation of osteocytes on osteoclastogenesis in wear debris-induced osteolysis. Results: Using Dmp1-cre conditional Cx43 knockout mice and the MLO-Y4 osteocyte cell line, we demonstrated that Cx43 deficiency reduced bone resorption and osteoclastogenesis, thereby improving bone remodeling in a Ti particle-induced osteolysis model. Sequencing analysis revealed that Cx43 gene expression changes might be linked to oxidative stress and the Janus Kinase (JAK)-STAT pathway. Elevated Cx43 expression in osteocytes stimulated by Ti particles increased STAT1 protein phosphorylation, induced oxidative stress, elevated the Receptor Activator of Nuclear Factor Kappa-Β Ligand (RANKL)/Osteoprotegerin (OPG) ratio, and promoted osteoclast activation and bone resorption. Conversely, Cx43 gene knockout decreased STAT1 protein phosphorylation and enhanced Nuclear Factor Erythroid 2-Related Factor 2 (NrF2) protein expression. Blocking the JAK-STAT signaling pathway activated by Cx43 increased NrF2 expression, reduced reactive oxygen species levels, and subsequently decreased the RANKL/OPG ratio. Innovation and Conclusions: This study identified a novel mechanism where Cx43 in osteocytes promoted osteoclastogenesis through JAK-STAT pathway activation and oxidative stress in wear debris-induced osteolysis. These findings highlighted the critical role of Cx43 in bone resorption and suggested targeting Cx43 or the JAK-STAT pathway as potential therapeutic strategies to mitigate osteolysis and improve implant longevity. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Jiawei Ouyang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Chai
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Rheumatology and Immunology, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Chunguang Sun
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shendong Wang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chang She
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Xu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
42
|
Xu Y, Sui Y, Jiang R, Wang X, Suda M, Niimi M, Mao Z, Zhang Z, Zhang SL, Fan J, Yao J. Sulfhydrated albumin transmits H 2S signaling and ameliorates DOX-induced multiorgan injuries. Redox Biol 2025; 83:103631. [PMID: 40228337 PMCID: PMC12018206 DOI: 10.1016/j.redox.2025.103631] [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: 03/10/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025] Open
Abstract
Hydrogen sulfide (H2S) is a vital signaling molecule involved in various physiological processes; however, the mechanisms underlying its systemic signaling remain poorly understood. We hypothesized that albumin, the predominant plasma protein and a vital sulfhydryl carrier, mediated systemic H2S signaling, which could potentially treat H2S-deficient diseases. This study aimed to investigate this hypothesis. Our results showed the presence of sulfhydrated proteins in normal mouse serum, with albumin being particularly enriched. The level of sulfhydration was influenced by H2S availability and the redox environment. In vitro incubation of albumin with NaHS resulted in an increased number of sulfhydrated groups. Under reductive conditions, this sulfhydrated albumin (-SSH-Alb) released substantial amounts of H2S. When -SSH-Alb was added to cultured endothelial cells, it activated the cAMP signaling pathway, upregulated cystathionine γ-lyase (CSE) expression, and enhanced intracellular H2S levels. In an in vitro inflammatory model involving macrophages and endothelial cells, -SSH-Alb inhibited macrophage adhesion, reduced LPS-induced expression of adhesion molecules, and suppressed cytokine production and inflammasome activation. These effects correlated with improved cellular redox status. Furthermore, in vivo administration of -SSH-Alb protected mice from doxorubicin (DOX)-induced cardiotoxicity and intestinal damage. It improved mouse mortality, and alleviated ferroptotic cardiac injury and gut barrier dysfunction. These therapeutic benefits were associated with rebalanced local and systemic redox status. In summary, our study reveals that -SSH-Alb reserves, transmits, and amplifies H2S signals and exhibits significant anti-inflammatory and antioxidant properties. This characteristic of -SSH-Alb holds promise for preventing and treating a wide range of diseases.
Collapse
Affiliation(s)
- Yijun Xu
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Yang Sui
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Rui Jiang
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Xin Wang
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Mika Suda
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Manabu Niimi
- Division of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Zhimin Mao
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Zhen Zhang
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Shao-Ling Zhang
- Department of Medicine, Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, H2 X 0A9, Montréal, QC, Canada
| | - Jianglin Fan
- Division of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan; Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China.
| | - Jian Yao
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan.
| |
Collapse
|
43
|
Song S, Liu H, Geng X, Chen R, Zheng R, Li N, Chen W, Wei W, Ren J. Assessment of the participation rate and effectiveness of upper gastrointestinal cancer screening in China: a multicenter population-based screening study. Endoscopy 2025. [PMID: 40199480 DOI: 10.1055/a-2564-9756] [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] [Indexed: 04/10/2025]
Abstract
This study estimated the participation rate and effectiveness of endoscopic screening of upper gastrointestinal cancer (UGIC) in China.From 2014 to 2015, we recruited 192805 adults from seven cities in China to build a prospective cohort for UGIC screening, including esophageal cancer and gastric cancer. Overall, 39991 participants were at high risk for UGIC according to an established risk score system and were recommended for endoscopic examination. Participation rates were reported, and their associated factors were explored. After inverse probability of treatment weighting (IPTW) based on propensity scores, Poisson regression was used to estimate the incidence rate ratio (IRR) and 95%CI for UGIC incidence and mortality between screened and non-screened groups from cohort entry until 31 December 2021.10442 participants underwent endoscopy, giving a participation rate of 26.1%. Higher participation was observed among individuals with higher education levels, history of upper gastrointestinal diseases, and family history of cancer. After a median follow-up of 6.8 years (interquartile range 6.7-6.9), UGIC incidences after IPTW were 94.6 per 100000 person-years in the screened group and 84.7 per 100000 person-years in the non-screened group. The weighted UGIC mortality rates were 13.1 and 33.4 per 100 000 person-years, respectively. The screened group had significantly lower UGIC mortality than the non-screened group, with a weighted IRR of 0.39 (95%CI 0.19-0.82).Participation in endoscopic screening among a high-risk UGIC population in China was low. We found that one-time endoscopic screening significantly reduced mortality caused by UGIC.
Collapse
Affiliation(s)
- Song Song
- Office of Cancer Screening, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han Liu
- Office of Cancer Screening, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianyi Geng
- Office of Cancer Screening, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ru Chen
- National Central Cancer Registry, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rongshou Zheng
- National Central Cancer Registry, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ni Li
- Office of Cancer Screening, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenqiang Wei
- National Central Cancer Registry, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiansong Ren
- Office of Cancer Screening, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Gu H, Yu W, Feng P, Zeng C, Cao Q, Chen F, Wang Z, Shen H, Wu Y, Wang S. Circular RNA circSTX12 regulates osteo-adipogenic balance and proliferation of BMSCs in senile osteoporosis. Cell Mol Life Sci 2025; 82:149. [PMID: 40192802 PMCID: PMC11977094 DOI: 10.1007/s00018-025-05684-y] [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: 06/20/2024] [Revised: 01/03/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
Increased adipogenic differentiation and decreased osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) along with slow self-renewal are pivotal causes for decreased bone formation in senile osteoporosis. Circular RNAs (circRNAs) play important roles in cell proliferation and differentiation, and are closely related to osteoporosis. Whether circRNAs orchestrate the adipo-osteogenic balance and the proliferation of BMSCs in osteoporosis remains unclear. We found in this study that circSTX12 was abnormally upregulated in bone sections from osteoporosis patients and in BMSCs from aged mice, as well as in later-generation human BMSCs in culture. Knockdown of circSTX12 in BMSCs resulted in enhanced osteogenesis, decreased adipogenesis, and increased proliferation capacity; circSTX12 overexpression had the opposite effect. RNA pull-down and mass spectrometry revealed the interactions between circSTX12 with CBL and LMO7. At the molecular level, circSTX12 regulated cell fate in BMSCs by competitively binding to CBL, reducing the ubiquitination-mediated degradation of MST1 and thereby activating the Hippo pathway, a key regulator of adipo-osteogenic balance. Knockdown of circSTX12 promoted the nuclear localization of YAP. In addition, our findings suggest that LMO7 mediates circSTX12-induced BMSCs proliferation by regulating the transcription of CCNA2, CCNH, and CCND1. In vivo, injection of antisense oligonucleotides (ASOs) to knockdown circSTX12 promoted bone formation in aged mice. Our results provide evidence for circSTX12 as a regulator of adipo-osteogenic differentiation and proliferation of BMSCs through binding to CBL and LMO7, respectively. Targeting circSTX12 may be a novel approach for osteoporosis treatment.
Collapse
Affiliation(s)
- Huimin Gu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Wenhui Yu
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Pei Feng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Chenying Zeng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Qian Cao
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Fenglei Chen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Ziming Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Huiyong Shen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
| | - Yanfeng Wu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
| | - Shan Wang
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
| |
Collapse
|
45
|
Zhang YL, Song HB, Xue YW. Machine learning-based reconstruction of prognostic staging for gastric cancer patients with different differentiation grades: A multicenter retrospective study. World J Gastroenterol 2025; 31:104466. [PMID: 40248057 PMCID: PMC12001173 DOI: 10.3748/wjg.v31.i13.104466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND The prognosis of gastric cancer (GC) patients is poor, and an accurate prognostic staging system would help assess patients' prognostic status before treatment and determine appropriate treatment strategies. AIM To develop positive lymph node ratio (LNR) and machine learning (ML)-based staging systems for GC patients with varying differentiation. METHODS This multicenter retrospective cohort study included 11772 GC patients, with 5612 in the training set (Harbin Medical University Cancer Hospital) and 6160 in the validation set (Surveillance, Epidemiology, and End Results Program database). X-tile software identified optimal cutoff values for the positive LNR, and five ML models were developed using pT and LNR staging. Risk scores were divided into seven stages, constructing new staging systems tailored to different tumor differentiation levels. RESULTS In both the training and validation sets, regardless of the tumor differentiation level, LNR staging demonstrated superior prognostic stratification compared to pN. Extreme Gradient Boosting exhibited better predictive performance than the other four models. Compared to tumor node metastasis staging, the new staging systems, developed for patients with different degrees of differentiation, showed significantly better predictive performance. CONCLUSION The new positive lymph nodes ratio staging and integrated staging systems constructed for GC patients with different differentiation grades exhibited better prognostic stratification capabilities.
Collapse
Affiliation(s)
- Yong-Le Zhang
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Hai-Bin Song
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Ying-Wei Xue
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| |
Collapse
|
46
|
Zhang H, Sun N, Li F, Wang Q, Sun Z, Zhang Y, Wang L, Zhao C, Fu Y. Construction of a modified TNM staging system and prediction model based on examined lymph node counts for gastric cancer patients at pathological stage N3. Front Oncol 2025; 15:1569736. [PMID: 40248208 PMCID: PMC12003143 DOI: 10.3389/fonc.2025.1569736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/13/2025] [Indexed: 04/19/2025] Open
Abstract
Background Examined lymph node (ELN) count is a critical factor affecting the number of metastatic lymph nodes (MLNs). The impact of the ELN number on survival and staging remains unclear. Methods This study included 4,291 stage N3 GC patients from the SEER database (training cohort) and 567 stage N3 GC patients from the FAHZZU database (validation cohort). The optimal ELN count and stage migration were investigated, and a modified TNM (mTNM) staging system including the ELN count was proposed. LASSO regression and random forest analyses were used to screen and evaluate the variables associated with survival, and an mTNM-based nomogram was constructed. The performance of the mTNM staging system and mTNM-based nomogram were compared with that of the 8th edition of the TNM staging system. Results The optimal threshold of the ELN count was identified as 21. An insufficient number of ELNs (≤ 21) was associated with poorer survival outcomes and led to stage migration in all N3 patients. A new mTNM staging system was proposed, integrating the ELN count into the TNM staging system (8th edition). LASSO regression analysis revealed that age, tumor size, adjuvant chemotherapy, adjuvant radiotherapy, and the mTNM system were associated with overall survival (OS) outcomes, and random forest analysis revealed that the mTNM system was the most important variable for predicting survival. An mTNM-based nomogram was constructed to predict 1-, 3-, and 5-year OS rates. Compared with the TNM staging system (8th edition), the mTNM staging system and mTNM-based nomogram showed superior prognosis discriminative ability, better predictive accuracy, and greater net improvement in survival outcomes. Conclusions The optimal ELN count for N3 GC patients was 21. The mTNM staging system and mTNM-based nomogram showed superior discriminative ability, predictive accuracy, and greater net benefit for OS outcomes.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Nan Sun
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feng Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiyang Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhao Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yawei Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunlin Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
47
|
Gao F, Li J. Identification of ferroptosis-related gene signatures in temporal lobe epilepsy with hippocampal sclerosis. Front Neurosci 2025; 19:1530182. [PMID: 40242460 PMCID: PMC11999976 DOI: 10.3389/fnins.2025.1530182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/07/2025] [Indexed: 04/18/2025] Open
Abstract
Background Ferroptosis is a form of regulated cell death that damages neurons in the central nervous system. In this study, we aimed to construct ferroptosis-related gene signatures in temporal lobe epilepsy with hippocampal sclerosis (TLE-HS) and explore their diagnostic role in TLE-HS. Methods The GSE205661 dataset was acquired for training purposes, while the GSE71058 was obtained to serve as the validation dataset. Subsequently, ferroptosis-related differentially expressed genes (FR-DEGs) in TLE-HS were further analyzed. We used weighed gene co-expression network analysis (WGCNA) algorithm, single-factor logistic regression analysis, and LASSO algorithm to screen characteristic FR-DEGs. Then, the receiver operating characteristic (ROC) was used to evaluate the value of these characteristic genes in disease diagnosis. Finally, a long non-coding RNA (lncRNA)-microRNA (miRNA)-messenger RNA (mRNA) network was constructed. Results We identified 141 FR-DEGs in TLE-HS, and these genes were enriched in T-cell activation involved in immune response and signaling pathways related to lipids and atherosclerosis. Further WGCNA was performed to select 47 overlapping FR-DEGs, which were significantly enriched in 13 biological processes and 14 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, including the negative regulation of apoptotic process and ferroptosis. Four genes, namely PDK4, SMPD1, GPT2, and METTL14, were identified as signature genes in TLE-HS. Moreover, the ROC derived from the four genes in GSE205661 and GSE71058 for predicting TLE-HS had an area under the curve (AUC) of 0.988 and 0.929, respectively. Furthermore, the lncRNA-miRNA-mRNA network constructed from the 4 FR-DEGs consisted of 5 lncRNAs and 14 miRNAs. The signatures based on four FR-DEGs were found to be a strong predictor of TLE-HS, and they may represent valuable therapeutic targets for TLE-HS.
Collapse
Affiliation(s)
| | - Jinzi Li
- Department of Pediatrics, Yanbian University Hospital, Yanji, China
| |
Collapse
|
48
|
Tufail M, Jiang CH, Li N. Wnt signaling in cancer: from biomarkers to targeted therapies and clinical translation. Mol Cancer 2025; 24:107. [PMID: 40170063 PMCID: PMC11963613 DOI: 10.1186/s12943-025-02306-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/15/2025] [Indexed: 04/03/2025] Open
Abstract
The Wnt signaling pathway plays a crucial role in development and tissue homeostasis, regulating key cellular processes such as proliferation, differentiation, and apoptosis. However, its abnormal activation is strongly associated with tumorigenesis, metastasis, and resistance to therapy, making it a vital target for cancer treatment. This review provides a comprehensive insight into the role of Wnt signaling in cancer, examining its normal physiological functions, dysregulation in malignancies, and therapeutic potential. We emphasize the importance of predicting Wnt signaling sensitivity and identify key biomarkers across various cancer types. Additionally, we address the challenges and future prospects of Wnt-targeted therapies, including biomarker discovery, advancements in emerging technologies, and their application in clinical practice.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China.
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
49
|
Chen Q, Liu S, Liu Y, Liu H, Wang H, Guo L, Xu H, Guo X, Wang X, Kang R, Zheng L, Zhang S. Lifetime risk of developing and dying from cancer in Henan Province, China: current status, temporal trends, and disparities. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:140-148. [PMID: 40265089 PMCID: PMC12010400 DOI: 10.1016/j.jncc.2024.11.005] [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: 04/01/2024] [Revised: 07/17/2024] [Accepted: 11/25/2024] [Indexed: 04/24/2025] Open
Abstract
Objective To understand the current status and changing trends in the lifetime risk of residents in Henan Province, China to develop and die from cancer. Methods Lifetime risk was estimated using the Adjusted for Multiple Primaries (AMP) method, incorporating cancer incidence, mortality, and all-cause mortality data from 55 cancer registries in Henan Province, China. Estimates were calculated overall and stratified by gender and area. The annual percent change (APC) in lifetime risk from 2010 to 2020, stratified by gender and cancer site, was estimated using a log-linear model. Results In 2020, the lifetime risk of developing and dying from cancer was 30.19 % (95 % CI: 29.63 %-30.76 %) and 23.62 % (95 % CI: 23.28 %-23.95 %), respectively. These estimates were higher in men, with values of 31.22 % (95 % CI: 30.59 %-31.85 %) for developing cancer and 26.73 % (95 % CI: 26.29 %-27.16 %) for dying from cancer, compared with women, who had values of 29.02 % (95 % CI: 28.12 %-29.91 %) and 20.08 % (95 % CI: 19.51 %-20.64 %), respectively. There were also geographical differences, with higher estimates in urban areas compared with rural areas. Residents had the highest lifetime risk of developing lung cancer, with a rate of 6.94 %, followed by breast cancer (4.14 %), stomach cancer (3.95 %), esophageal cancer (3.75 %), and liver cancer (2.86 %). Similarly, the highest lifetime risk of dying from cancer was observed for the following sites: lung (5.99 %), stomach (3.60 %), esophagus (3.39 %), liver (2.78 %), and colorectum (1.55 %). Overall, the lifetime risk of developing cancer increased, with an APC of 0.75 % (P < 0.05). Varying trends were observed across different cancer sites. There were gradual decreases in nasopharynx, esophagus, stomach, and liver cancers. Conversely, increasing trends were noted for most other sites, with the highest APCs observed in thyroid, prostate, lymphoma, kidney, and gallbladder cancers. Conclusion The lifetime risks of developing and dying from cancer were 30.19 % and 23.62 %, respectively. Variations in cancer risk across different regions, genders, specific cancer sites, and over calendar years provide important information for cancer prevention and policy making in the population.
Collapse
Affiliation(s)
- Qiong Chen
- Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Shuzheng Liu
- Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Yin Liu
- Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Hongwei Liu
- Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Hong Wang
- Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Lanwei Guo
- Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Huifang Xu
- Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Xiaoli Guo
- Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Xiaoyang Wang
- Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Ruihua Kang
- Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Liyang Zheng
- Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Shaokai Zhang
- Department of Cancer Epidemiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| |
Collapse
|
50
|
Galli E, Patelli G, Villa F, Gri N, Mazzarelli C, Mangoni I, Sgrazzutti C, Ghezzi S, Sartore-Bianchi A, Belli LS, De Carlis L, Vanzulli A, Siena S, Bencardino K. Circulating blood biomarkers for minimal residual disease in hepatocellular carcinoma: A systematic review. Cancer Treat Rev 2025; 135:102908. [PMID: 40058162 DOI: 10.1016/j.ctrv.2025.102908] [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: 11/09/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 04/08/2025]
Abstract
BACKGROUND Relapse after radical treatment remains a major concern in hepatocellular carcinoma (HCC), affecting 50-75 % of early-stage cases within 5 years. Early recurrence prediction is a clinical unmet need. Circulating blood biomarkers could provide a minimally invasive approach to detect minimal residual disease (MRD) post-intervention. Although alpha-fetoprotein has been the primary biomarker in this setting, its MRD sensitivity is limited to 50-70 %. This systematic review aims to summarize available evidence regarding the clinical validity and potential utility of emerging circulating blood biomarkers for MRD detection in HCC patients. METHODS We searched PubMed and Embase for peer-reviewed articles and abstracts published up to 2025, and ClinicalTrials.gov for ongoing trials on circulating blood biomarkers for MRD in HCC. RESULTS A total of 91 studies (74 with results and 17 ongoing, out of 2,386) were retrieved. We evaluated various blood biomarkers, including circulating DNA (cDNA, N = 24), circulating tumor cells (CTCs, N = 20), circulating RNA (cRNA, N = 8), and other miscellaneous (N = 22) for MRD detection in HCC. These biomarkers demonstrated encouraging results, albeit with notable heterogeneity. In particular, circulating tumor DNA (ctDNA) and CTCs stand as the most robust novel approaches, with 50-80 % sensitivity and specificity up to 94 %. Nonetheless, none of the 17 ongoing studies involve biomarker-driven intervention to prove clinical utility. CONCLUSIONS Novel circulating blood biomarkers are mature for MRD detection in HCC. However, variability in methodologies and results highlights the need for further validation. We encourage the investigation of CTCs and/or ctDNA in interventional trials to assess clinical utility. This biomarker-driven approach may enhance adjuvant treatment effectiveness in MRD-positive cases while minimizing toxicity in MRD-negative patients.
Collapse
Affiliation(s)
- Edoardogregorio Galli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giorgio Patelli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy.
| | - Federica Villa
- Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Nicole Gri
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Chiara Mazzarelli
- Hepatology and Gastroenterology Unit, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Iacopo Mangoni
- Department of General Surgery and Transplantation, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Silvia Ghezzi
- Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Division of Clinical Research and Innovation, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Luca Saverio Belli
- Hepatology and Gastroenterology Unit, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Luciano De Carlis
- Department of General Surgery and Transplantation, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Angelo Vanzulli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Department of Radiology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Katia Bencardino
- Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|