1
|
Sakamoto T, Akiyama S, Narasaka T, Tuchiya K. Advancements and limitations of image-enhanced endoscopy in colorectal lesion diagnosis and treatment selection: A narrative review. DEN OPEN 2026; 6:e70141. [PMID: 40353217 PMCID: PMC12061549 DOI: 10.1002/deo2.70141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/23/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality, highlighting the need for early detection and accurate lesion characterization. Traditional white-light imaging has limitations in detecting lesions, particularly those with flat morphology or minimal color contrast with the surrounding mucosa. It also struggles to distinguish neoplastic from non-neoplastic lesions. These limitations led to the development of image-enhanced endoscopy (IEE). Image-enhanced endoscopy modalities such as narrow-band imaging, blue laser imaging, linked color imaging, and texture and color enhancement imaging enhance mucosal surface and vascular pattern visualization, thereby improving lesion detection and characterization. In contrast, red dichromatic imaging is primarily designed to enhance the visibility of deep blood vessels, making it particularly useful during therapeutic endoscopies, such as identifying bleeding sources and monitoring post-treatment hemostasis. Although IEE enhances lesion detection and characterization, it remains limited in assessing submucosal invasion depth, which is a key factor in treatment decisions. Endoscopic submucosal dissection requires accurate prediction of invasion depth; however, IEE mainly reflects superficial features. Endoscopic ultrasound and artificial intelligence-assisted diagnostics have emerged as complementary techniques for improving depth assessment and lesion classification. Additionally, IEE plays a critical role in detecting ulcerative colitis-associated neoplasia (UCAN), which often presents with a flat morphology and indistinct borders. High-definition chromoendoscopy and IEE modalities enhance detection; however, inflammation-related changes limit diagnostic accuracy. Artificial intelligence and molecular biomarkers may improve UCAN diagnosis. This review examines the role of IEE in lesion detection and treatment selection, its limitations, and complementary techniques such as endoscopic ultrasound and artificial intelligence. We also explored pit pattern diagnosis using crystal violet staining and discussed emerging strategies to refine colorectal cancer screening and management.
Collapse
Affiliation(s)
- Taku Sakamoto
- Division of GastroenterologyUniversity of Tsukuba HospitalIbarakiJapan
| | - Shintaro Akiyama
- Division of GastroenterologyUniversity of Tsukuba HospitalIbarakiJapan
| | - Toshiaki Narasaka
- Division of GastroenterologyUniversity of Tsukuba HospitalIbarakiJapan
| | - Kiichiro Tuchiya
- Division of GastroenterologyUniversity of Tsukuba HospitalIbarakiJapan
| |
Collapse
|
2
|
Duizer C, Salomons M, van Gogh M, Gräve S, Schaafsma FA, Stok MJ, Sijbranda M, Kumarasamy Sivasamy R, Willems RJL, de Zoete MR. Fusobacterium nucleatum upregulates the immune inhibitory receptor PD-L1 in colorectal cancer cells via the activation of ALPK1. Gut Microbes 2025; 17:2458203. [PMID: 39881579 PMCID: PMC11784648 DOI: 10.1080/19490976.2025.2458203] [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: 05/13/2024] [Revised: 01/02/2025] [Accepted: 01/14/2025] [Indexed: 01/31/2025] Open
Abstract
Fusobacterium nucleatum is a Gram-negative oncobacterium that is associated with colorectal cancer. The molecular mechanisms utilized by F. nucleatum to promote colorectal tumor development have largely focused on adhesin-mediated binding to the tumor tissue and on the pro-inflammatory capacity of F. nucleatum. However, the exact manner in which F. nucleatum promotes inflammation in the tumor microenvironment and subsequent tumor promotion remains underexplored. Here, we show that both living F. nucleatum and sterile F. nucleatum-conditioned medium promote CXCL8 release from the intestinal adenocarcinoma HT-29 cell line. We determined that the observed pro-inflammatory effect was ALPK1-dependent in both HEK293 and HT-29 cells and that the released F. nucleatum molecule had characteristics that match those of the pro-inflammatory ALPK1 ligand ADP-heptose or related heptose phosphates. In addition, we determined that not only F. nucleatum promoted an ALPK1-dependent pro-inflammatory environment but also other Fusobacterium species such as F. varium, F. necrophorum and F. gonidiaformans generated similar effects, indicating that ADP-heptose or related heptose phosphate secretion is a conserved feature of the Fusobacterium genus. By performing transcriptional analysis of ADP-heptose stimulated HT-29 cells, we found several inflammatory and cancer-related pathways to be differentially regulated, including DNA mismatch repair genes and the immune inhibitory receptor PD-L1. Finally, we show that stimulation of HT-29 cells with F. nucleatum resulted in an ALPK1-dependent upregulation of PD-L1. These results aid in our understanding of the mechanisms by which F. nucleatum can affect tumor development and therapy and pave the way for future therapeutic approaches.
Collapse
Affiliation(s)
- Coco Duizer
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Moniek Salomons
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Merel van Gogh
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sanne Gräve
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Freke A. Schaafsma
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maaike J. Stok
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Merel Sijbranda
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Rob J. L. Willems
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel R. de Zoete
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
3
|
Liu Z, Zhang H, Wang J, Yao Y, Wang X, Liu Y, Fang W, Liu X, Zheng Y. Clca1 deficiency exacerbates colitis susceptibility via impairment of mucus barrier integrity and gut microbiota homeostasis. Microbiol Res 2025; 297:128191. [PMID: 40300372 DOI: 10.1016/j.micres.2025.128191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
The intestinal mucus barrier has emerged as a promising therapeutic target for inflammatory bowel disease. Understanding its regulatory mechanisms is critical for elucidating ulcerative colitis (UC) pathogenesis, improving diagnostics, guiding treatments, and preventing relapse. Chloride Channel Accessory 1 (Clca1), a constituent of the mucus layer, remains understudied in colitis. Here, we investigated Clca1's role in mucosal immunity and intestinal homeostasis using experimental colitis models. Clca1-deficient (Clca1-/-) mice displayed compromised mucus layer integrity, reduced neutrophil infiltration, and gut microbiota dysbiosis. Notably, Clca1-/- mice exhibited exacerbated colitis severity following dextran sulfate sodium (DSS) challenge, accompanied by a diminished goblet cell populations. Fecal microbiota transplantation (FMT) studies revealed that gut microbiota critically modulates divergent phenotypic outcomes between genotypes. Our findings establish Clca1 as a multifunctional regulator of mucus barrier integrity through mechanisms involving goblet cell maintenance, neutrophil-mediated immunity, and host-microbiota crosstalk. These results advance the understanding of UC pathogenesis and identify Clca1-associated pathways as potential targets for barrier restoration therapies.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hong Zhang
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jingjing Wang
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yutong Yao
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaoyi Wang
- Core Facility Center, The First Afliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yang Liu
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Xingyin Liu
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Biochemistry, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.
| | - Yi Zheng
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| |
Collapse
|
4
|
Song T, Zhuang M, Wong ZYD, Xu G, Tang ML, Kou B, Sun X. Discovery of a deuterated TNF-α small molecule modulator for potential treatment of ulcerative colitis. Eur J Med Chem 2025; 291:117616. [PMID: 40239485 DOI: 10.1016/j.ejmech.2025.117616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Tumor necrosis factor-alpha (TNF-α) is an important target for the treatment of inflammatory diseases. Targeting TNF-α inhibition, such as antibody drug infliximab and adalimumab, has emerged as an effective therapeutic strategy for managing the most difficult-to-treat chronic ulcerative colitis (UC). So far, there are no small molecule TNF-α inhibitors available on the market for the treatment of UC. Previously, we reported an indanone analogue (R)-STU104 showed considerable inhibitory activity on TNF-α production in both acute and chronic mouse models of UC with a favorable safety profile. However, further development potential of this compound was greatly limited due to its poor metabolic stability in human liver microsomes and suboptimal pharmacokinetic profiles in mice. Herein, we discovered a deuterated TNF-α small molecule modulator (R)-104-6D-01, which demonstrated promising clinical potential for the treatment of UC. This new compound exhibited enhanced oral bioavailability and exposure in pharmacokinetic studies, as well as superior anti-UC efficacy in a DSS-induced mouse UC model, compared with (R)-STU104 at a dosage of 30 mg/kg/d. Collectively, (R)-104-6D-01 proves to be a promising candidate of potential use in treating UC as an oral TNF-α small molecule modulator.
Collapse
Affiliation(s)
- Tong Song
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | - Mengxiao Zhuang
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | | | - Ge Xu
- Jiangsu Angeltech Pharmaceuticals Co., Ltd, 4F, 19 South Taiping Road, Chengxiang Town, Taicang City, Suzhou, Jiangsu Province, 215400, China
| | - Mei-Lin Tang
- Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Buyu Kou
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | - Xun Sun
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China; The Institutes of Integrative Medicine of Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China.
| |
Collapse
|
5
|
Ding H, Tang X, Tang W. Effect of adjuvant chemotherapy with toad venom injection in the treatment of intermediate and advanced colon cancer and its effect on cellular immunity, PTEN, and PI3k. Anticancer Drugs 2025; 36:495-500. [PMID: 39964699 DOI: 10.1097/cad.0000000000001706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
The objective of this study is to explore the effect of adjuvant chemotherapy with toad venom injection in patients with intermediate and advanced colon cancer, in order to provide new reference drugs for clinical treatment. Prospectively, 148 patients with mid-stage to late-stage colon cancer in our hospital from January 2021 to May 2023 were selected for the study and randomly divided into two groups of 74 cases each. The control group was treated with FOLFOX4 chemotherapy, and the observation group was treated with four consecutive chemotherapy cycles based on the control group combined with toad venom injection. The treatment effects, adverse reactions, quality of life improvement rate, prognosis and cellular immune indexes [natural killer (NK) cells, CD4 + /CD8 + , CD4 + , CD3 + ], phosphatase tensin gene ( PTEN ), phosphatidylinositol-3-kinase (PI3k), and serine threonine protein kinase (pAKT) protein expression before and after treatment were counted in the two groups. The total effective rate of treatment in the observation group was 58.11% (43/74) after four cycles of chemotherapy, which was higher than that in the control group of 41.89% (31/74) ( P < 0.05). After two cycles of chemotherapy and four cycles of chemotherapy, PTEN , CD4 + /CD8 + , CD4 + , CD3 + , and NK cells in peripheral blood were higher in the observation group than in the control group, and PI3k and pAKT were lower than in the control group ( P < 0.05). There was no statistically significant difference in the rate of adverse reactions in the observation group compared with the control group ( P > 0.05); the improvement rate of quality of life in the observation group was better than that in the control group after four chemotherapy cycles of treatment ( P < 0.05); the survival rate was 75.00% (54/72) in the observation group compared with 54.29% (38/70) in the control group at 1-year follow-up. Toad venom injection adjuvant chemotherapy is effective in treating patients with intermediate and advanced colon cancer, which can upregulate PTEN level, inhibit PI3k and AKT expression, and improve immune function and quality of life of patients, thus improving prognosis.
Collapse
Affiliation(s)
- Haijun Ding
- General Surgery, Tianshan Branch of Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine
| | - Xuedian Tang
- General Surgery, Tianshan Branch of Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine
| | - Wenjun Tang
- General Surgery, Shanghai Eighth People's Hospital, Shanghai, China
| |
Collapse
|
6
|
Fang X, Zhao J, Wu S, Liao P, Guan G. The intestinal toxicity mechanisms of triclosan and triclocarban and their possible clinical nutritional intervention mechanisms. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 376:126396. [PMID: 40345375 DOI: 10.1016/j.envpol.2025.126396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/20/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Triclosan (TCS) and triclocarban (TCC) are widely used as antimicrobial agents in personal care products. Their widespread use has become a potential environmental contaminant. This review reviews the mechanisms of intestinal toxicity of TCS and TCC and their potential nutritional intervention strategies. TCS and TCC can be metabolized to glucuronic acid conjugates in the host and subsequently uncoupled by microorganisms in the intestine to regenerate free forms of TCS and TCC. TCS and TCC are unique metabolic pathways that lead to accumulation in the gut, altering the structure of intestinal flora, increasing the relative abundance of pathogenic bacteria, while reducing the abundance of beneficial bacteria, thereby disrupting the balance of intestinal flora. In addition, they can interfere with the self-renewal and differentiation of ISCs, thereby weakening intestinal barrier function. TCS and TCC can also activate the TLR4-NFκB signaling pathway, inducing and exacerbating inflammatory responses. These mechanisms together lead to intestinal toxicity and have a significant negative impact on intestinal health. In order to cope with the intestinal toxicity caused by these mechanisms of action, this paper believes that prebiotics, probiotics, vitamins, minerals and herbal extracts can be used as potential nutritional interventions to reduce the intestinal toxicity of TCS and TCC by regulating intestinal microbiota, enhancing intestinal barrier function and inhibiting inflammatory response. Although preliminary studies have shown the potential benefits of these interventions, their specific efficacy and safety still need further study.
Collapse
Affiliation(s)
- Xinyu Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China; Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, 410125, China
| | - Jinfeng Zhao
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China; Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, 410125, China
| | - Simin Wu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China; Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, 410125, China
| | - Peng Liao
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, 410125, China; Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, 410219, China.
| | - Guiping Guan
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China.
| |
Collapse
|
7
|
Li FL, Wang BB, Zeng KF, Chen HY, Wu XH, Wang Y, Lin HC, Li WL, Zhao XD. Exploring the Anticancer Effects of Xianliu Jieduan Fang on Colitis-Associated Colorectal Cancer Through Network Pharmacology and Experimental Validation. Biomed Chromatogr 2025; 39:e70102. [PMID: 40324811 DOI: 10.1002/bmc.70102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/15/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
This study evaluated the therapeutic effects of Xianliu Jieduan Fang (XLJDF) on colitis-associated colorectal cancer (CAC) and explored its molecular mechanisms through network pharmacology and experimental validation. Using an AOM/DSS-induced CAC mouse model, we evaluated XLJDF's efficacy. Active components were identified by UHPLC-QE-HRMS. Targets were predicted using SwissTargetPrediction and PubChem, while disease genes were obtained from GeneCards, DisGeNET, and TTD. Core targets and pathways were analyzed via Cytoscape and Metascape. Mechanisms were validated through molecular docking and experiments. XLJDF improved colon pathology and identified 68 active compounds, including nine key components like Kaempferol and Luteolin. Network analysis revealed 959 targets with 29 core genes (AKT1, CTNNB1, GSK3B, etc.). KEGG analysis showed XLJDF primarily acts through Wnt signaling, regulating apoptosis and cell migration. Experimental validation confirmed XLJDF inhibits Wnt/β-catenin pathway by preventing GSK3β inactivation. XLJDF exerts anti-CAC effects via a multi-component, multi-target network. Our study identifies key active compounds and demonstrates that XLJDF suppresses the Wnt/β-catenin pathway by preventing GSK3β inactivation, thereby inhibiting β-catenin stabilization.
Collapse
Affiliation(s)
- Fang-Lan Li
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Bei-Bei Wang
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Ke-Feng Zeng
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hao-Yang Chen
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xi-Hua Wu
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yun Wang
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Hong-Cheng Lin
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Wei-Lin Li
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xiang-Dong Zhao
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
8
|
Wang C, Liang W, Zhong J, Liu J, Zhou C, Ma C, Liao Y, Gao Y, Zhao J, He Y. m6A-mediated regulation of CPSF6 by METTL3 promotes oxaliplatin resistance in colorectal cancer through enhanced glycolysis. Cell Signal 2025; 130:111676. [PMID: 40010558 DOI: 10.1016/j.cellsig.2025.111676] [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: 10/02/2024] [Revised: 12/16/2024] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Oxaliplatin resistance poses a significant challenge in colorectal cancer (CRC) treatment. Recent studies have implicated CPSF6 in cancer progression and drug resistance, although its role in chemotherapy resistance and regulatory mechanisms is unclear. This study investigates CPSF6's involvement in oxaliplatin resistance in CRC and its regulation via m6A methylation by METTL3. We assessed CPSF6 expression in oxaliplatin-resistant (OxR) CRC cell lines (HT29-OxR and HCT116-OxR) compared to sensitive counterparts using qRT-PCR and Western blotting. CPSF6 was significantly upregulated in OxR cells, and its knockdown reduced cell viability, colony formation, and glycolytic activity while increasing apoptosis. m6A modification of CPSF6 mRNA was elevated in OxR cells, correlating with increased METTL3 expression. METTL3 knockdown decreased CPSF6 levels and m6A enrichment, enhancing mRNA degradation, while its overexpression stabilized CPSF6 mRNA, promoting resistance. Xenograft experiments showed that CPSF6 knockdown suppressed tumor growth and glycolysis. Thus, CPSF6 is identified as a mediator of oxaliplatin resistance in CRC, regulated by the METTL3/m6A axis, suggesting potential therapeutic targets to overcome resistance.
Collapse
Affiliation(s)
- Chengxing Wang
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Weijun Liang
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Jietao Zhong
- Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China; Department of Gastroenterology, Jiangmen Central Hospital, Guangdong 529000, China
| | - Jiachen Liu
- Department of Nuclear Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangdong 510000, China
| | - Chaorong Zhou
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Changyi Ma
- Department of Radiology, Jiangmen Central Hospital, Guangdong 529000, China
| | - Yuehua Liao
- Department of Pathology, Jiangmen Central Hospital, Guangdong 529000, China
| | - Yuan Gao
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Jinglin Zhao
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China.
| | - Yaoming He
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China.
| |
Collapse
|
9
|
Luo Y, Lu F, Luo L, Li Y. Plasma fibrinogen levels towards cancer incidence: a systematic review and meta-analysis of epidemiological studies. Discov Oncol 2025; 16:844. [PMID: 40397214 PMCID: PMC12095739 DOI: 10.1007/s12672-025-02691-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 05/12/2025] [Indexed: 05/22/2025] Open
Abstract
IMPORTANCE Understanding the association between fibrinogen levels and cancer incidence is crucial for elucidating potential diagnostic and therapeutic implications in oncology. OBJECTIVE This meta-analysis aimed to comprehensively evaluate the relationship between circulating fibrinogen levels and various tumour types. REGISTRATION This systematic review had been registered in PROSPERO (ID: CRD42024616015). DATA SOURCES A systematic review search was performed on PUBMED, EMBASE and Cochrane databases until June 3, 2023. STUDY SELECTION Studies that fulfilled our pre-established inclusion criteria were incorporated into our analysis. These criteria encompassed prospective cohort, case-control, and nested case-control designs, all featuring histopathologically confirmed primary cancers. Furthermore, we included studies that had reported odds ratios (ORs), relative ratios (RRs) or hazard ratios (HRs) along with their corresponding 95% confidence intervals (95% CIs), ensuring the reliability and comparability of the data across studies. DATA EXTRACTION AND SYNTHESIS Two independent authors meticulously extracted data from eligible studies, ensuring rigour and accuracy. Subsequently, we performed statistical analyses using the robust STATA version 12.0 software, guaranteeing the reliability of our findings. Moreover, we carried out in-depth subgroup analyses, categorizing studies based on tumour type, to meticulously explore and quantify the variations in the correlation between fibrinogen levels and tumour incidence. Through this approach, we have gained a nuanced understanding of the potential heterogeneity of this relationship across different cancer types. MAIN OUTCOMES AND MEASURES The major outcome of this study centred on elucidating the relationship between fibrinogen levels and cancer incidence, with subsequent subgroup analyses conducted to delve deeper into this relationship within specific tumour types. This approach aimed to provide a comprehensive understanding of how fibrinogen levels may varying influence different cancer types, thereby offering potential insights into the identification of novel risk factors or biomarkers for further clinical investigation. RESULTS Twelve studies were meticulously incorporated into the meta-analysis. Notably, significant heterogeneity was observed across these studies, necessitating careful interpretation of the results. The meta-analysis demonstrated a compelling connection between elevated fibrinogen levels and an increased risk of cancer incidence, with an hazard ratio (HR) of 1.33 (95% CI: 1.16, 1.51; p = 0.000), demonstrating a statistically significant finding. Further subgroup analyses delved into specific cancer types and identified significant associations with smoking-related cancer (HR = 1.79; 95% CI: 1.54, 2.09; p = 0.000), lung cancer (HR = 1.98; 95% CI: 1.62, 2.43; p = 0.000) and colorectal cancer (HR = 1.27; 95% CI: 1.00, 1.62; p = 0.048). These findings underline the potential significance of fibrinogen levels as a potential biomarker or risk factor for these particular cancer types. CONCLUSIONS AND RELEVANCE Elevated plasma fibrinogen levels have been significantly associated with an increased incidence of cancer, particularly in the cases of lung and colorectal cancers. These compelling findings underscore the potential value of fibrinogen levels as a diagnostic or prognostic biomarker in cancer management. Accordingly, further studies and clinical validation are urgently needed to fully explore the role of fibrinogen in cancer and its potential application in clinical practice.
Collapse
Affiliation(s)
- Yuexin Luo
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei Province, China
- First Clinic School, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Furong Lu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medicial College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Lihua Luo
- Department of Oncology, The Central Hospital of Enshi Autonomous Prefecture, Enshi Clinical College of Wuhan University, No. 234, Aviation Avenue, Enshi Tujia and Miao Autonomous Prefecture, 445000, Hubei Province, China.
| | - Yuting Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei Province, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei Province, China.
- Hubei Key Laboratory of Precision Radiation Oncology, No. 1277 Jiefang Avenue, Jianghan District, Wuhan City, 430022, Hubei Province, China.
| |
Collapse
|
10
|
Chen X, Zhang J, Gao F, Liu N, Du H, Li J, Li Z, Chen R. Exercise therapy: an effective approach to mitigate the risk of cancer metastasis. World J Surg Oncol 2025; 23:192. [PMID: 40380237 PMCID: PMC12082867 DOI: 10.1186/s12957-025-03846-7] [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: 11/29/2024] [Accepted: 05/06/2025] [Indexed: 05/19/2025] Open
Abstract
Cancer metastasis is a primary contributor to cancer-related mortality, and mitigating the risk of metastasis has emerged as a central concern in oncology research. In recent years, exercise therapy, as a non-pharmacological intervention, has received considerable attention for its ability to enhance patients' quality of life and prognosis. Exercise significantly inhibits cancer spread, diminishes cancer risk, and improves therapy outcomes. Nonetheless, the exact mechanisms via which exercise inhibits the dissemination and metastasis of cancer cells are not fully elucidated. This review seeks to examine the mechanisms and prospective research avenues of exercise treatment in mitigating the risk of cancer metastasis. Moreover, it methodically examines pertinent clinical and scientific data, along with the efficacy of exercise therapies in real-world applications. The evaluation moreover suggests future research avenues, including a more profound exploration of mechanisms, the augmentation of clinical trials, the advancement of personalized and precision exercise therapy, and enhanced multidisciplinary collaboration. Exercise therapy shows significant potential in mitigating the risk of cancer metastasis, and its incorporation into holistic cancer treatment frameworks is advised to improve patients' general health and prognostic results.
Collapse
Affiliation(s)
- Xiaoyan Chen
- Department of Physical Therapy, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
- Medical College of Nanchang Institute of Technology, Nanchang, 330044, China
| | - Junfeng Zhang
- Department of Physical Therapy, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Feng Gao
- Department of Physical Therapy, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Na Liu
- The Hong Kong Polytechnic University Faculty of Health and Social Sciences, Kowloon, HK, China
| | - Huijun Du
- Department of Physical Therapy, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Jiuhu Li
- Department of Physical Therapy, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Zhi Li
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
- Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
| | - Rong Chen
- School of Physical Education, Sports Fitness Research Center, East China Jiaotong University, Nanchang, 330013, China.
| |
Collapse
|
11
|
Yaker L, Saliba J, Scott LPC, Sood AK, Gujral P, Orozco-Alonso E, Yan X, Yeh A, Blank V. NFE2L3 regulates inflammation and oxidative stress-related genes in the colon. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119987. [PMID: 40360021 DOI: 10.1016/j.bbamcr.2025.119987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 04/04/2025] [Accepted: 05/07/2025] [Indexed: 05/15/2025]
Abstract
The molecular mechanisms leading to inflammatory bowel disease (IBD) are only partially understood. We investigated the role of the transcription factor NFE2L3 in a mouse model of colitis by inducing inflammation using dextran sodium sulfate (DSS). We confirmed the presence of inflammation by histological analysis and elevated levels of the inflammation marker lipocalin-2 (LCN2) in the stool. We found that Lcn2 transcript levels are significantly less elevated in Nfe2l3-/- mice than wild type mice. We further showed a reduction of Nfe2l3 mRNA, in wildtype mice upon DSS treatment. We cross referenced ENCODE ChIP data of NFE2L3 binding partners MAFF and MAFK with known IBD and DSS effectors and identified Stat1, Hmox1, and Slc7a11 as potential NFE2L3 targets. These proteins are induced during colitis to suppress the immune response, reduce oxidative stress, and trigger ferroptosis, respectively. We analyzed the candidate targets and observed an increase in their protein expression upon DSS treatment in wild type but not in Nfe2l3-/- mice. Furthermore, in the absence of DSS, we observed an increase in the basal levels of pSTAT1 and SLC7A11 proteins in Nfe2l3-/- mice. These data suggest that the NFE2L3 transcription factor primes the microenvironment towards a pro-inflammatory ready state during inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Linda Yaker
- Lady Davis Institute for Medical Research, Montreal, H3T 1E2, Canada; Department of Medicine, McGill University, Montreal, H4A 3J1, Canada
| | - James Saliba
- Lady Davis Institute for Medical Research, Montreal, H3T 1E2, Canada; Department of Medicine, McGill University, Montreal, H4A 3J1, Canada
| | - Liam P C Scott
- Lady Davis Institute for Medical Research, Montreal, H3T 1E2, Canada; Department of Medicine, McGill University, Montreal, H4A 3J1, Canada
| | - Anantpreet Kaur Sood
- Lady Davis Institute for Medical Research, Montreal, H3T 1E2, Canada; Department of Medicine, McGill University, Montreal, H4A 3J1, Canada
| | - Palak Gujral
- Lady Davis Institute for Medical Research, Montreal, H3T 1E2, Canada; Department of Medicine, McGill University, Montreal, H4A 3J1, Canada
| | - Eduardo Orozco-Alonso
- Lady Davis Institute for Medical Research, Montreal, H3T 1E2, Canada; Department of Medicine, McGill University, Montreal, H4A 3J1, Canada
| | - Xingyue Yan
- Lady Davis Institute for Medical Research, Montreal, H3T 1E2, Canada; Department of Medicine, McGill University, Montreal, H4A 3J1, Canada
| | - Adam Yeh
- Lady Davis Institute for Medical Research, Montreal, H3T 1E2, Canada; Department of Medicine, McGill University, Montreal, H4A 3J1, Canada
| | - Volker Blank
- Lady Davis Institute for Medical Research, Montreal, H3T 1E2, Canada; Department of Medicine, McGill University, Montreal, H4A 3J1, Canada; Department of Physiology, McGill University, Montreal, QC, H3G 1Y6, Canada.
| |
Collapse
|
12
|
Zhang C, Zhang N, Zhang Y, Gao X, Liu X, Qiu B, Qi S, Li Y, Li L, Liu W. Effects of Lactobacillus plantarum-loaded W1/O/W2 emulsions stabilized by ultrasonically treated pea protein on DSS-induced colitis in mice. Int J Biol Macromol 2025; 311:143956. [PMID: 40348251 DOI: 10.1016/j.ijbiomac.2025.143956] [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: 12/12/2024] [Revised: 04/23/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
This study evaluates the efficacy of Lactobacillus plantarum (L. plantarum) NBRC15891 encapsulated in a W1/O/W2 emulsion stabilized by ultrasonically treated pea protein nanoparticles (HIU-PPI-NP) for alleviating colitis. The results indicate that HIU-PPI-NP effectively preserved bacterial viability during simulated gastrointestinal digestion, with a survival rate of 72.62 %, significantly higher than that of free bacteria (0.0017 %). In vivo, the L. plantarum-loaded emulsion alleviated dextran sodium sulfate (DSS) -induced colitis in mice by improving body weight, reducing the disease activity index (DAI), and decreasing myeloperoxidase (MPO) activity in the colonic tissue. Gene expression analysis revealed reduction in inflammation markers (TNF-α, IL-6) and restoration of MUC2 expression. Furthermore, microbiota analysis indicated that L. plantarum-loaded emulsion altered the microbiota composition by promoting beneficial genera, such as Lachnospiraceae_NK4A136_group and Eubacterium_siraeum_group, while inhibiting the pathogenic growth of Escherichia-Shigella. Additionally, an increase in short-chain fatty acid (SCFA) concentrations was observed in mice treated with the L. plantarum-loaded emulsion. Importantly, neither free L. plantarum cells nor L. plantarum-free emulsion exhibited comparable therapeutic effects on colitis, suggesting that this emulsion system could aid in protecting L. plantarum and enhancing its efficacy. These findings underscore the potential of HIU-PPI-NP-stabilized emulsion as a novel probiotic delivery system for gut health application.
Collapse
Affiliation(s)
- Chao Zhang
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Nan Zhang
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China; College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Yu Zhang
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Xueyan Gao
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xia Liu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Bin Qiu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Shasha Qi
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Yonghao Li
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Lingfei Li
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| | - Wei Liu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China.
| |
Collapse
|
13
|
Li G, Ma J, Wu L, Zhang H, Lin Y, Xu H, Gu M, Li K, Dong H, Huang Y, Wu H. Moxibustion regulates KDM4D expression and modulates lipid metabolism to inhibit tumor proliferation in CAC mice. Cancer Cell Int 2025; 25:173. [PMID: 40325472 PMCID: PMC12054041 DOI: 10.1186/s12935-025-03798-8] [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: 01/10/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Lysine demethylase 4D (KDM4D) and aberrant lipid metabolism are implicated in the development and progression of colitis-associated cancer (CAC). Moxibustion, a therapeutic approach in traditional Chinese medicine, can inhibit intestinal inflammation and improve the intestinal mucosa. METHODS Mice were intraperitoneally injected with AOM, and three cycles of 3-2-2% DSS-free drinking water were administered to establish a CAC mouse model. Moxibustion and KDM4D inhibitor 5-c-8HQ intervention were performed for 30 days after modeling was completed. IHC staining was used to observe the expression of the nuclear-associated antigen Ki67 (Ki67), proliferating cell nuclear antigen (PCNA), and IL-33 in the colon. The expression of colon KDM4D and β-Catenin was observed by immunofluorescence staining and RT‒qPCR. LC‒MS pseudotargeted metabolomic sequencing was used to semiquantitatively detect the expression levels of lipids. RESULTS Moxibustion inhibited the proliferation of colon tumors in CAC mice, improved histopathology, and reduced the expression of PCNA and Ki67 in the colon. Using kdm4d knockout technology, it was initially confirmed that kdm4d is a key gene affecting CAC tumor proliferation. The inhibition of colon tumor proliferation in CAC mice by moxibustion is associated with the suppression of abnormal activation of the colon KDM4D/β-Catenin signaling pathway. LC-MS-targeted metabolomics revealed abnormal lipid metabolism in the colons of CAC mice. Moxibustion may affect the cholinergic metabolism pathway in the colon of CAC mice and regulate lipids such as sphingomyelin SM (d18:1/26:0) and triacylglycerol TAG58:7 (18:0). After kdm4d knockout, lipid disorders in the colons of CAC mice were partially restored. The kdm4d gene may be involved in the mechanism underlying the effect of moxibustion on lipid metabolism in the CAC colon. CONCLUSIONS Moxibustion inhibited the proliferation of colon tumors in CAC mice, inhibited the activation of the tumor-promoting signaling pathway KDM4D/β-Catenin, and improved lipid metabolism disorders in the colon, thus providing a promising strategy for the clinical adjuvant treatment of colorectal cancer.
Collapse
Affiliation(s)
- Guona Li
- Yueyang Hospital of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, 650 Wanping Road, Shanghai, 200030, China
| | - Jindan Ma
- Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 568 Xinhua Road, Shanghai, 200052, China
| | - Luyi Wu
- Yueyang Hospital of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Hanxiao Zhang
- Yueyang Hospital of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Yaying Lin
- Yueyang Hospital of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Hongxiao Xu
- Yueyang Hospital of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Muen Gu
- Yueyang Hospital of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Kunshan Li
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, 650 Wanping Road, Shanghai, 200030, China
| | - Hongsheng Dong
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, 650 Wanping Road, Shanghai, 200030, China.
| | - Yan Huang
- Yueyang Hospital of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China.
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, 650 Wanping Road, Shanghai, 200030, China.
| | - Huangan Wu
- Yueyang Hospital of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China.
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, 650 Wanping Road, Shanghai, 200030, China.
| |
Collapse
|
14
|
Ayari A, Jedidi S, Dakhli N, Sammari H, Dhawefi N, Sebai H. Fresh Beetroot Juice Alleviates Combined Ulcerative Colitis and Constipation by Restoring Physiological and Biochemical Balances in a Murine Model. Neurogastroenterol Motil 2025:e70064. [PMID: 40317625 DOI: 10.1111/nmo.70064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/12/2025] [Accepted: 04/18/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Beetroot (Beta vulgaris L.) is well known for its medicinal uses, particularly in managing gastrointestinal disorders. This study investigates the protective effects of fresh red beet juice (FBRJ) on gastrointestinal complications caused by co-administration of dextran sulfate sodium (DSS) and loperamide (LOP), which induce ulcerative colitis and constipation, respectively. METHODS Adult rats were divided into groups and subjected to a 7-day treatment with 5% DSS to induce ulcerative colitis, followed by LOP (3 mg/kg, body weight [b.w.]) for 7 days to cause constipation. FBRJ (5 and 10 mL/kg, b.w.) or yohimbine (YOH) (2 mg/kg, b.w.) was administered 1 h after LOP each day for 7 days. Therapeutic outcomes were evaluated based on macroscopic and histological changes in the gastrointestinal tract, gastric emptying, gastrointestinal transit, oxidative stress parameters, and inflammatory markers. KEY RESULTS FBRJ significantly alleviated gastrointestinal dysfunctions caused by DSS and/or LOP, improving both gastric emptying and gastrointestinal transit in a dose-dependent manner (p < 0.05). Specifically, compared with the ulcerative/constipated group, the animals treated with the FBRJ showed a significant increase (52.43% ± 4.65% to 66.23% ± 6.78%) of gastric emptying (GE) andgastrointestinal transit (GIT: 48.08% ± 3.32% to 62.46% ± 4.98%) in a dose-dependent manner. It also modulated antioxidant defense systems by inducing enzyme activities and reducing lipid peroxidation, which had been significantly disrupted by the combined effects of DSS and LOP. Furthermore, inflammatory markers, including C-reactive protein (CRP), pro-inflammatory cytokines, and white blood cell counts, were significantly reduced in both plasma and colonic mucosa. CONCLUSIONS AND INTERFERENCES We suggest that FBRJ significantly protects against DSS-induced colitis and LOP-induced constipation, involving several mechanisms such as increasing secretion and peristaltic activity, reducing inflammation, and preserving the antioxidant properties.
Collapse
Affiliation(s)
- Ala Ayari
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), Higher Institute of Biotechnology of Béja, University of Jendouba, Beja, Tunisia
| | - Saber Jedidi
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), Higher Institute of Biotechnology of Béja, University of Jendouba, Beja, Tunisia
- National Institute of Technologies and Sciences of Kef (INTeK), University of Jendouba, El Kef, Tunisia
| | - Nouha Dakhli
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), Higher Institute of Biotechnology of Béja, University of Jendouba, Beja, Tunisia
| | - Houcem Sammari
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), Higher Institute of Biotechnology of Béja, University of Jendouba, Beja, Tunisia
| | - Nourhène Dhawefi
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), Higher Institute of Biotechnology of Béja, University of Jendouba, Beja, Tunisia
| | - Hichem Sebai
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), Higher Institute of Biotechnology of Béja, University of Jendouba, Beja, Tunisia
| |
Collapse
|
15
|
León-Vega II, Oregon R, Schnoor M, Vadillo E. From Ulcerative Colitis to Metastatic Colorectal Cancer: The Neutrophil Contribution. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:814-830. [PMID: 39889826 DOI: 10.1016/j.ajpath.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
Ulcerative colitis (UC) is an inflammatory colon and rectum disease affecting approximately 5 million people worldwide. There is no cure for UC, and approximately 8% of patients with UC develop colorectal cancer (CRC) by gradual acquisition of mutations driving the formation of adenomas and their progression to adenocarcinomas and metastatic disease. CRC constitutes 10% of total cancer cases worldwide and 9% of cancer deaths. Both UC and CRC have an increasing incidence worldwide. Although the epithelium has been well studied in UC and CRC, the contribution of neutrophils is less clear. Neutrophils are rapidly recruited in excessive amounts from peripheral blood to the colon during UC, and their overactivation in the proinflammatory UC tissue environment contributes to tissue damage. In CRC, the role of neutrophils is controversial, but emerging evidence suggests that their role depends on the evolution and context of the disease. The role of neutrophils in the transition from UC to CRC is even less clear. However, recent studies propose neutrophils as therapeutic targets for better clinical management of both diseases. This review summarizes the current knowledge on the roles of neutrophils in UC and CRC.
Collapse
Affiliation(s)
- Iliana I León-Vega
- Department of Molecular Biomedicine, Cinvestav-National Polytechnic Institute, Mexico City, Mexico
| | - Reyna Oregon
- Oncology Research Unit, Oncology Hospital, National Medical Center, Mexican Institute of Social Security, Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine, Cinvestav-National Polytechnic Institute, Mexico City, Mexico.
| | - Eduardo Vadillo
- Oncology Research Unit, Oncology Hospital, National Medical Center, Mexican Institute of Social Security, Mexico City, Mexico.
| |
Collapse
|
16
|
Zhang H, Zhang N, Yang X, Wang C, Yang Q, Luo J, Ye T. BRAF mutation cancer, colorectal cancer, tumor associated lymph node structure and immune microenvironment study: MAPK protein kinase molecular action and SIRPG-CD47 protein signaling pathway. Int J Biol Macromol 2025; 307:142191. [PMID: 40101830 DOI: 10.1016/j.ijbiomac.2025.142191] [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: 01/14/2025] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 03/20/2025]
Abstract
BRAF mutation affects the biological characteristics and microenvironment of the tumor during the development of colorectal cancer. Tumor-associated lymph nodes are the key sites of immune response. This study aimed to systematically evaluate the impact of BRAF gene mutations on the remodeling of the CRC immune microenvironment, with a particular focus on their effects on the maturation and function of TLS·In this study, clinical samples of CRC patients were collected, and immune cell subsets were analyzed by single-cell RNA sequencing, and pseudo-temporal locus analysis and spatial transcriptome analysis were performed to explore intercellular communication and functional enrichment analysis. The distribution and maturity of TLS were evaluated by immunohistochemistry and multiple fluorescence staining techniques, and statistical analysis was performed.The results showed that BRAF mutation significantly affected the number and maturity of lymphatic structures infiltrated by tumors, and was negatively correlated with patient prognosis. BRAF mutations lead to alterations in T cell subsets, particularly the dual role of CD4+ CXCL13 cells in TLS maturation. B-cell subpopulation analysis revealed functional deficits in CRC patients with BRAF mutations, which further drove the remodeling of the tumor immune microenvironment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Oncology, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Nenglin Zhang
- Department of Gastroenterology, First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China
| | - Xiaodi Yang
- Department of Oncology, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Chen Wang
- Department of Oncology, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Qinghui Yang
- Department of Oncology, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Jing Luo
- Department of Oncology, Minhang Hospital, Fudan University, Shanghai 201199, China; Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, China.
| | - Tao Ye
- Department of Oncology, Minhang Hospital, Fudan University, Shanghai 201199, China; Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, China.
| |
Collapse
|
17
|
Pawlak M, Kałuzińska-Kołat Ż, Pasieka ZW, Kołat D, Płuciennik E. The critical role of COL1A1 revealed by integrated bioinformatics analysis of differentially-expressed genes in colorectal cancer and inflammatory bowel disease. Comput Biol Med 2025; 190:110116. [PMID: 40179807 DOI: 10.1016/j.compbiomed.2025.110116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE There is an urgent need to identify biomarkers of tumorigenesis for colitis-associated cancer (CAC) as early cancer detection remains crucial for patients with inflammatory bowel disease (IBD). This in silico study examines the relationship between IBD and CAC, with particular regard to differentially-expressed genes (DEGs). METHODS Integrated bioinformatics tools and public databases were employed. Data from GEO (GSE102133, GSE48958, GSE9348, GSE83687, GSE138202) were processed using GEOexplorer. DEGs were then functionally annotated with DAVID, SRplot, and integrated analysis via Metascape. Validation used Oncopression and Human Protein Atlas. Survival analysis employed GEPIA2. miRNA interactions were studied via miRTargetLink 2.0. Immune infiltration was analyzed with TIMER 2.0. COL1A1 expression and mutations were examined using cBioPortal, Kaplan-Meier plotter, and DNA methylation was analyzed using MethSurv. Correlation of COL1A1 gene promoter methylation with tissue type and clinical data was performed using the UALCAN database. The ROC analysis of COL1A1 was conducted in the R environment. RESULTS Our analysis identified three potential hub genes (ICAM1, LAMC1, and COL1A1), which are overexpressed in IBD and cancer tissues compared to normal tissue, and hence may play a role in CAC. Furthermore, patients with lower COL1A1 expression had longer disease-free survival (p = 0.01) than those with higher expression. Therefore, this gene was chosen for further analysis and identified as the most crucial. CONCLUSION COL1A1 reveals significant immunohistochemistry, mutations, and methylation data. Further studies involving machine learning and clinical data are required to validate the results.
Collapse
Affiliation(s)
- Martyna Pawlak
- Department of Biomedical Sciences, Faculty of Medicine, Medical University of Lodz, Poland
| | - Żaneta Kałuzińska-Kołat
- Department of Functional Genomics, Medical University of Lodz, Poland; Department of Biomedicine and Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Poland
| | - Zbigniew W Pasieka
- Department of Biomedicine and Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Poland
| | - Damian Kołat
- Department of Functional Genomics, Medical University of Lodz, Poland; Department of Biomedicine and Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Poland
| | | |
Collapse
|
18
|
Hayashi R, Ueno N, Watanabe H, Kobayashi Y, Sakatani A, Takahashi K, Yuzawa S, Ando K, Tani C, Kashima S, Shonaka T, Moriichi K, Tanabe H, Tanino M, Fujiya M. Unresectable Ulcerative Colitis Associated Colon Cancer in a Young Japanese Patient. Intern Med 2025; 64:1344-1349. [PMID: 39370253 DOI: 10.2169/internalmedicine.4160-24] [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] [Indexed: 10/08/2024] Open
Abstract
We herein present the case of a 30-year-old Japanese male patient with ulcerative colitis (UC) who was admitted to our hospital because of significant ascites. Upon evaluation, the patient was diagnosed with unresectable UC-associated colon cancer (UCAC), localized in the transverse colon. Using gene profiling of the tumor tissue, anti-epidermal growth factor receptor (EGFR) antibody combination chemotherapy was selected. Subsequently, the patient exhibited a temporary response to this regimen, with an enhancement in his quality of life and he was able to survive for 12 months. This case underscores the potential benefits of aggressive chemotherapy tailored to the gene profile in UCAC treatment, offering insights into potential avenues for improving the patient prognosis.
Collapse
Affiliation(s)
- Ryunosuke Hayashi
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
| | - Nobuhiro Ueno
- Department of General Medicine, Asahikawa Medical University Hospital, Japan
- Department of Gastroenterological Sciences, Asahikawa Medical University, Japan
| | - Hiromu Watanabe
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
| | - Yu Kobayashi
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
| | - Aki Sakatani
- Department of Gastroenterological Sciences, Asahikawa Medical University, Japan
| | - Keitaro Takahashi
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
| | - Sayaka Yuzawa
- Department of Diagnostic Pathology, Asahikawa Medical University Hospital, Japan
| | - Katsuyoshi Ando
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
| | - Chikayoshi Tani
- Division of Gastrointestinal Surgery, Department of Surgery, Asahikawa Medical University, Japan
| | - Shin Kashima
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
| | - Tatsuya Shonaka
- Division of Gastrointestinal Surgery, Department of Surgery, Asahikawa Medical University, Japan
| | - Kentaro Moriichi
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
| | - Hiroki Tanabe
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
| | - Mishie Tanino
- Department of Diagnostic Pathology, Asahikawa Medical University Hospital, Japan
| | - Mikihiro Fujiya
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
- Department of Gastroenterological Sciences, Asahikawa Medical University, Japan
| |
Collapse
|
19
|
East JE, Gordon M, Nigam GB, Sinopoulou V, Bateman AC, Din S, Iacucci M, Kabir M, Lamb CA, Wilson A, Al Bakir I, Dhar A, Dolwani S, Faiz O, Hart A, Hayee B, Healey C, Leedham SJ, Novelli MR, Raine T, Rutter MD, Shepherd NA, Subramanian V, Vance M, Wakeman R, White L, Trudgill NJ, Morris AJ. British Society of Gastroenterology guidelines on colorectal surveillance in inflammatory bowel disease. Gut 2025:gutjnl-2025-335023. [PMID: 40306978 DOI: 10.1136/gutjnl-2025-335023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/12/2025] [Indexed: 05/02/2025]
Abstract
Patients with inflammatory bowel disease (IBD) remain at increased risk for colorectal cancer and death from colorectal cancer compared with the general population despite improvements in inflammation control with advanced therapies, colonoscopic surveillance and reductions in environmental risk factors. This guideline update from 2010 for colorectal surveillance of patients over 16 years with colonic inflammatory bowel disease was developed by stakeholders representing UK physicians, endoscopists, surgeons, specialist nurses and patients with GRADE (Grading of Recommendations Assessment, Development and Evaluation) methodological support.An a priori protocol was published describing the approach to three levels of statement: GRADE recommendations, good practice statements or expert opinion statements. A systematic review of 7599 publications, with appraisal and GRADE analysis of trials and network meta-analysis, where appropriate, was performed. Risk thresholding guided GRADE judgements.We made 73 statements for the delivery of an IBD colorectal surveillance service, including outcome standards for service and endoscopist audit, and the importance of shared decision-making with patients.Core areas include: risk of colorectal cancer, IBD-related post-colonoscopy colorectal cancer; service organisation and supporting patient concordance; starting and stopping surveillance, who should or should not receive surveillance; risk stratification, including web-based multivariate risk calculation of surveillance intervals; colonoscopic modalities, bowel preparation, biomarkers and artificial intelligence aided detection; chemoprevention; the role of non-conventional dysplasia, serrated lesions and non-targeted biopsies; management of dysplasia, both endoscopic and surgical, and the structure and role of the multidisciplinary team in IBD dysplasia management; training in IBD colonoscopic surveillance, sustainability (green endoscopy), cost-effectiveness and patient experience. Sixteen research priorities are suggested.
Collapse
Affiliation(s)
- James Edward East
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Morris Gordon
- School of Medicine, University of Central Lancashire, Preston, UK
| | - Gaurav Bhaskar Nigam
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | - Adrian C Bateman
- Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, Southampton, Hampshire, UK
| | - Shahida Din
- The Edinburgh IBD Unit, Western General Hospital, Edinburgh, UK
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Marietta Iacucci
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Misha Kabir
- Division of Gastrointestinal Services, University College Hospitals NHS Trust, London, UK
| | - Christopher Andrew Lamb
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Ana Wilson
- Department of Gastroenterology, St Mark's Hospital and Academic Institute, London, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Ibrahim Al Bakir
- Gastroenterology Department, Chelsea and Westminster Hospital, London, UK
| | - Anjan Dhar
- Department of Gastroenterology, Darlington Memorial Hospital, Darlington, Durham, UK
- Teesside University, Middlesbrough, UK
| | - Sunil Dolwani
- Division of Population Medicine, School of Medicine, Cardiff University, Cardiff, UK
| | - Omar Faiz
- Department of Surgery and Cancer, Imperial College London, London, UK
- Department of Colorectal Surgery, St Mark's Hospital and Academic Institute, London, UK
| | - Ailsa Hart
- Department of Gastroenterology, St Mark's Hospital and Academic Institute, London, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Bu'Hussain Hayee
- King's Health Partners Institute for Therapeutic Endoscopy, King's College Hospital NHS Foundation Trust, London, UK
| | - Chris Healey
- Department of Gastroenterology, Airedale NHS Foundation Trust, Keighley, West Yorkshire, UK
| | - Simon John Leedham
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Stem Cell Biology Lab, Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Marco R Novelli
- Department of Histopathology, University College London, London, UK
| | - Tim Raine
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, Cambridgeshire, UK
| | - Matthew D Rutter
- Department of Gastroenterology, University Hospital of North Tees, Stockton-on-Tees, UK
| | - Neil A Shepherd
- Gloucestershire Cellular Pathology Laboratory, Cheltenham General Hospital, Cheltenham, Gloucestershire, UK
| | - Venkataraman Subramanian
- Department of Gastroenterology, St James's University Hospital, Leeds, UK
- Division of Gastroenterology and Surgical Sciences, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Margaret Vance
- Department of Gastroenterology, St Mark's Hospital and Academic Institute, London, UK
| | | | - Lydia White
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Nigel J Trudgill
- Department of Gastroenterology, Sandwell and West Birmingham NHS Trust, West Bromwich, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - A John Morris
- Department of Gastroenterology, Glasgow Royal Infirmary, Glasgow, UK
| |
Collapse
|
20
|
Santerre A, Huizar-López MDR, Coronilla-Martínez J, Ortiz-Lazareno PC, Casas-Solís J. Lacticaseibacillus casei 393 modulates KRAS and APC expression and cytokine levels in colitis-associated colon cancer. J Gastrointest Oncol 2025; 16:568-579. [PMID: 40386603 PMCID: PMC12078812 DOI: 10.21037/jgo-24-667] [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: 09/18/2024] [Accepted: 02/26/2025] [Indexed: 05/20/2025] Open
Abstract
Background Colitis-associated colon cancer (CAC) is a specific subset of colorectal cancer (CRC) affecting patients with inflammatory bowel diseases (IBDs). Chronic colon inflammation orchestrates immune surveillance or escape and may drive neoplastic initiation and progression. Lacticaseibacillus casei 393 (L. casei 393) is a lactic acid microorganism that, beyond its nutritional value, provides health benefits. To explore the therapeutic potential of this probiotic against CAC, we evaluated colon histopathology, circulating cytokines, and the expression of the Kristen rat viral sarcoma oncogene homolog (KRAS) and the adenomatosis polyposis coli (APC) tumor-suppressing gene in the murine model of CAC induced with azoxymethane (AOM) and dextran sodium sulfate (DSS). Methods BALB/c mice (n=7/group) received two doses of AOM (10 mg/kg body weight) followed by three 5-day cycles of 2% DSS. L. casei 393 was administered orally [1×106 colony forming units (CFU)/100 µL/mouse/twice a week/6 months] either alone, before AOM-DSS, or starting at the same time as AOM-DSS. Colon histopathology was assessed by hematoxylin-eosin staining, circulating cytokines by flow cytometry, and the expression of colonic KRAS and APC by quantitative reverse transcription polymerase chain reaction (RT-qPCR). Results AOM-DSS induced CAC in BALB/c mice, which presented severe colon damage, high cytokine levels, and altered KRAS and APC expression. Conversely, L. casei 393 ingestions, starting at the same time as CAC induction, restored colon architecture and modulated cytokine levels and gene expression. Conclusions The present experimental work supports the therapeutic potential of L. casei 393 against CAC, as it shows that its ingestion restored the damaging effect of AOM-DSS through its anti-inflammatory properties that helped modulate KRAS and APC mRNA expression.
Collapse
Affiliation(s)
- Anne Santerre
- Cellular and Molecular Biology Department, University of Guadalajara, Zapopan, Mexico
| | | | | | - Pablo Cesar Ortiz-Lazareno
- Immunology Unit, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, Mexico
| | - Josefina Casas-Solís
- Cellular and Molecular Biology Department, University of Guadalajara, Zapopan, Mexico
| |
Collapse
|
21
|
Martínez-Ríos J, López-Pacheco CP, García-Zepeda EA, Soldevila G. CCR9 shapes the immune microenvironment of colorectal cancer modulating the balance between intratumoral CD8+ T cell and FoxP3+ Helios+ Treg subpopulations. PLoS One 2025; 20:e0321930. [PMID: 40305493 PMCID: PMC12043142 DOI: 10.1371/journal.pone.0321930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/12/2025] [Indexed: 05/02/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world and the second cause of death related to cancer. Regulatory T cell (Treg) infiltration is enriched in several tumor types including CRC and correlates with suppression of the anti-tumor immune response. In the large intestine, thymic Tregs (tTregs Helios+) and peripheral Tregs (pTregs Helios-) coexist and maintain intestinal homeostasis under steady state conditions. The recruitment of Treg cells to the intestine is orchestrated by the CCR9/CCL25 axis, which is potentiated under inflammatory conditions. Interestingly, the balance between cytotoxic CD8+ T cells and Tregs within the tumor microenvironment is critical for antitumor immunity and cancer progression. An elevated CD8+/Treg ratio has been associated with improved clinical outcomes in various cancers, including CRC. Therefore, here we investigate the potential role of chemokine receptor CCR9 on CD8+/Treg ratio and the effect of the recruitment of Treg subpopulations (Helios+ and Helios-) into the tumor microenvironment using the AOM/DSS induced colitis-associated colorectal cancer murine model. Our findings reveal that CCR9 deficiency leads to distinct alterations in the CRC microenvironment, characterized by decreased intratumoral Tregs Helios+. Also, the lack of the receptor leads to an improvement of the antitumor immune response, increasing the CD8+/Treg ratio within the tumor immune infiltrate. These results underscore the importance of CCR9 in shaping the immune microenvironment during CRC development.
Collapse
Affiliation(s)
- Jacobo Martínez-Ríos
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Cynthia Paola López-Pacheco
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Investigadora por México, Secretaría de Ciencia Tecnología y Humanidades (SECIHTI), Mexico City, Mexico
| | - Eduardo Alberto García-Zepeda
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gloria Soldevila
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio Nacional de Citometría de Flujo, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
22
|
Zhang X, Liu S, Xin R, Hu W, Zhang Q, Lu Q, Han L. Reactive oxygen species-responsive prodrug nanomicelle-functionalized Lactobacillus rhamnosus probiotics for amplified therapy of ulcerative colitis. MATERIALS HORIZONS 2025. [PMID: 40296848 DOI: 10.1039/d5mh00114e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Characterized by elevated reactive oxygen species (ROS) and disrupted gut flora, ulcerative colitis (UC) affects millions of patients worldwide. Probiotic therapy is commonly utilized in clinical practice to modulate intestinal flora and ameliorate colitis symptoms. Nonetheless, oral probiotics encounter challenges such as limited bioactivity, brief retention time, intricate pathological conditions, and singular efficacy. Here we designed plant-derived 18β-glycyrrhetinic acid (18β-GA) prodrug nanomicelles with ROS and inflammation-resolving capabilities, as well as anti-depressant effects, to protect probiotics and amplify their therapeutic effects in alleviating UC and UC-associated depression-like behaviors. Upon oral administration to UC lesion sites, prodrug nanomicelles can be dissociated by excessive ROS and release 18β-GA to attenuate colonic inflammatory responses and oxidative stress, which in turn provided a favorable microenvironment for LGG to repair intestinal barrier integrity and restore the gut microbiota. The synergistic therapeutic effects of STG nanomicelles and LGG alleviated UC-associated depression-like behavior by suppressing the activation of microglia and reducing neuroinflammation. This study introduces a promising strategy for oral nanomedicine with satisfactory therapeutic outcomes for the treatment of inflammatory diseases by integrating naturally derived small-molecule drugs with probiotics.
Collapse
Affiliation(s)
- Xinyue Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, Shandong, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266071, Shandong, China
| | - Shuyun Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, Shandong, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266071, Shandong, China
| | - Rui Xin
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, Shandong, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266071, Shandong, China
| | - Wenxiu Hu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, Shandong, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266071, Shandong, China
| | - Qiqi Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, Shandong, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266071, Shandong, China
| | - Qian Lu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, Shandong, China.
- Marine Traditional Chinese Medicine R&D Laboratory, Marine Biomedical Research Institute of Qingdao, Qingdao 266071, Shandong, China
| | - Lu Han
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, Shandong, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266071, Shandong, China
| |
Collapse
|
23
|
Li R, Liu Y, Liu Q, Guo Z, Wang B, Huang S, Wang Z, Liu F, Zhou Y, Wang P, Li T, Fu W, Han W. CMTM3 Promotes Colitis-associated Carcinogenesis Via CLTC Stabilization and Modulation of VE-cadherin. Cell Mol Gastroenterol Hepatol 2025:101528. [PMID: 40306490 DOI: 10.1016/j.jcmgh.2025.101528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND & AIMS Inflammatory bowel disease leads to increased risk of developing colitis-associated colon cancer (CAC). CMTM3 has a higher methylation level in colon cancer, and accumulating evidence suggests that chemokine-like factor-like MARVEL transmembrane domain-containing member 3 (CMTM3) participates in inflammation and cancer development. METHODS We explored the signs of azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced CAC in wild-type (WT) and Cmtm3 deficiency (Cmtm3-/-) mice. Experimental colitis was induced in Cmtm3-/- mice as well as mice with endothelial cell-specific deletion of Cmtm3. Disease phenotypes were investigated by body weight, disease activity index (DAI), colon length, histology, immune cell infiltration, and intestinal permeability. The mechanism was analyzed using bone marrow reconstitution, immunofluorescent staining, Western blot, immunoprecipitation, and pull-down experiments. RESULTS We found CMTM3 promoted CAC by aggravating colitis. Further, we revealed endothelial cell-specific deletion of Cmtm3 inhibited the colitis development. In vitro and in vivo mechanistic studies revealed that CMTM3 drove colitis by increasing clathrin-dependent downregulation of vascular endothelial-cadherin, thus causing vascular permeability. We further identified that CMTM3 interacted with clathrin heavy chain and inhibited clathrin heavy chain ubiquitination and proteasome-dependent degradation. Interestingly, Cmtm3 knockout and imatinib mesylate both targeted vascular permeability and had comparable efficacy. CONCLUSIONS Our study indicates that CMTM3 promotes CAC by aggravating colitis through causing vascular permeability, providing insights into targets for development of future therapies.
Collapse
Affiliation(s)
- Rongbin Li
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China
| | - Yuan Liu
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China
| | - Qiyao Liu
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China; Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine (BUCM), Institute of Liver Diseases, BUCM, Dongcheng District, Beijing, China
| | - Zixia Guo
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China
| | - Bingsu Wang
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China
| | - Sihua Huang
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China
| | - Zelin Wang
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China; School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, China; Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong, China
| | - Fujun Liu
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong, China
| | - Yifan Zhou
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
| | - Pingzhang Wang
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China
| | - Ting Li
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China
| | - Weiwei Fu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing, China.
| | - Wenling Han
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China.
| |
Collapse
|
24
|
Guo L, Li X. Nurse night shift work and risk of gastrointestinal cancers. Front Public Health 2025; 13:1532623. [PMID: 40356833 PMCID: PMC12066619 DOI: 10.3389/fpubh.2025.1532623] [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: 11/22/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
The prevalence of night-shift employment is on the rise among full-time and part-time workers globally. Those engaged in night-shift work encounter various biological challenges, including exposure to artificial light during nighttime and disruptions to their circadian rhythms. These factors, along with changes in daily routines and activities, may pose significant risks to the health of night workers. Notably, the number of individuals working overtime or on night shifts has increased across various sectors, particularly in transportation, healthcare, and manufacturing. The International Agency for Research on Cancer (IARC) has classified night-shift work as probably carcinogenic to humans (IARC Group 2A). Subsequent research has identified several potential mechanisms through which night-shift work may contribute to carcinogenicity: (1) disruption of circadian rhythms, (2) suppression of melatonin levels due to nighttime light exposure, (3) physiological alterations, (4) lifestyle changes, and (5) reduced vitamin D levels resulting from inadequate sunlight exposure. Colorectal cancer (CRC) poses a significant public health challenge, ranking as the second leading cause of cancer-related death worldwide in 2020. Other than CRC, other gastrointestinal cancers are also creating a great global health issue because of their morbidity and mortality rates. In this review, we highlight the role of night shifts in disturbing circadian rhythm and how this action leads to carcinogenesis in the GI tract.
Collapse
Affiliation(s)
- Lin Guo
- Medical Simulated Center, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiaojun Li
- School of Nursing, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
25
|
Yang L, Li H, Tang M, He L, Yang L. Circular RNAs in inflammatory bowel disease: a review of mechanisms, biomarkers and therapeutic potential. Front Immunol 2025; 16:1540768. [PMID: 40342413 PMCID: PMC12058709 DOI: 10.3389/fimmu.2025.1540768] [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: 12/06/2024] [Accepted: 04/02/2025] [Indexed: 05/11/2025] Open
Abstract
Inflammatory bowel disease (IBD) is an autoimmune disease of unknown etiology characterized by recurrent chronic inflammation of the gastrointestinal tract. The incidence of IBD is increasing and has become a focus of research on digestive diseases. Despite advances in understanding its multifactorial etiology, including genetic predisposition, microbiome dysbiosis, and immune dysregulation. However, the molecular mechanisms driving IBD pathogenesis remain incompletely elucidated. Circular RNA (circRNA) is a stable single-stranded RNA with a closed-loop structure and conserved nature. circRNA possesses multiple functions, such as adsorption of microRNAs and RNA-binding proteins, and is involved in the regulation of gene splicing and transcription, as well as protein translation. However, circRNAs in IBD progression and their clinical potential as biomarkers or therapeutic targets are yet to be systematically explored. In this review, we comprehensively synthesize recent advancements in circRNA research related to IBD, integrating evidence from in vitro, in vivo, and clinical studies. We systematically analyze aberrant circRNA expression profiles in IBD tissues (e.g., intestinal mucosa, peripheral blood, and exosomes) and discuss their mechanism of action contributions to inflammation, intestinal epithelial barrier dysfunction, autophagy, intestinal fibrosis, and colitis-associated cancer (CAC). Furthermore, we evaluate methodologies for circRNA detection and therapeutic modulation, including RNA interference, viral vector delivery, and PLGA MSs delivery system strategies. This review highlights the potential of circRNA-focused strategies in the diagnosis and treatment of IBD, offering a scientific foundation for advancing precision medicine in IBD management.
Collapse
Affiliation(s)
- Le Yang
- Department of Gastroenterology, Yiyang Central Hospital, Yiyang, China
| | - Huahui Li
- Institute of Biomedical and Health Engineering, Chinese Academy of Sciences Shenzhen Institutes of Advanced Technology, Shenzhen, China
| | - Min Tang
- Department of Pharmacy, Yiyang Medical College, Yiyang, China
| | - Lingnan He
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lijun Yang
- Department of Pharmacy, Yiyang Medical College, Yiyang, China
| |
Collapse
|
26
|
Wang J, Hao Y, Yang Y, Zhang Y, Xu C, Yang R. Gut microbiota derived indole-3-acetic acid ameliorates precancerous inflammatory intestinal milieu to inhibit tumorigenesis through IL-35. J Immunother Cancer 2025; 13:e011155. [PMID: 40274281 PMCID: PMC12020765 DOI: 10.1136/jitc-2024-011155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Gut microbiota can significantly alter the risk or progression of cancer by maintaining gut immune system homeostasis. However, the exact mechanism by which the gut microbiota and its metabolites influence colorectal tumorigenesis is unclear. METHODS The roles of tryptophan metabolite indole-3-acetic acid (IAA) in inflammation and tumor development were investigated in dextran sodium sulfate (DSS) and azoxymethane (AOM)-DSS mouse models with or without IAA supplementation and with or without Lactobacillus reuteri-produced IAA. Pregnane X receptor (PXR) knockout (KO) mice and aryl hydrocarbon receptor KO mice were used to explore the mechanism by which IAA regulates interleukin (IL)-35 expression. IL-35+ immune cells were stimulated in vitro and analyzed by flow cytometry. Additionally, metabolites were analyzed by liquid chromatography-mass spectrometry. RESULTS We found that IAA, a metabolite of tryptophan produced in the gut by L. reuteri, can inhibit the development of colitis by inducing IL-35 expression in immunosuppressant cells. HuREG3αIECtg mice had high levels of intestinal microbiota-derived IAA, and these mice were resistant to AOM-DSS-induced cancer. Patients with colorectal cancer also had low peripheral blood levels of IAA. Further studies revealed that IAA-producing L. reuteri alleviated colitis symptoms and inhibited colon tumors by inducing macrophages, T cells, and B cells to produce IL-35. Finally, PXR KO completely abolished the effects of IAA on immune cells. CONCLUSION We demonstrate that gut microbiota-derived IAA can improve the precancerous colon inflammatory environment through IL-35, thereby inhibiting tumorigenesis, suggesting that IAA may be a preventive factor for colitis-related cancers.
Collapse
Affiliation(s)
| | - Yang Hao
- Nankai University School of Medicine, Tianjin, China
| | - Yazheng Yang
- Nankai University School of Medicine, Tianjin, China
| | - Yuan Zhang
- Nankai University School of Medicine, Tianjin, China
| | - Chen Xu
- Nankai University, Tianjin, China
| | - Rongcun Yang
- Nankai University Medical School, Nankai University School of Medicine, Tianjin, China
| |
Collapse
|
27
|
Johnson B, Bath T, Huang X, Lamm M, Earles A, Eddington H, Dornisch AM, Jih LJ, Gupta S, Shah SC, Curtius K. Large language models for extracting histopathologic diagnoses of colorectal cancer and dysplasia from electronic health records. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.11.27.24318083. [PMID: 40313292 PMCID: PMC12045448 DOI: 10.1101/2024.11.27.24318083] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Background Accurate data resources are essential for impactful medical research, but available structured datasets are often incomplete or inaccurate. Recent advances in open-weight large language models (LLMs) enable more accurate data extraction from unstructured text in electronic health records (EHRs) but have not yet been thoroughly validated for challenging diagnoses such as inflammatory bowel disease (IBD)-related neoplasia. Objective Create a validated approach using LLMs for identifying histopathologic diagnoses in pathology reports from the nationwide Veterans Health Administration database, including patients with genotype data within the Million Veteran Program (MVP) biobank. Design Our approach utilizes simple 'yes/no' question prompts for following phenotypes of interest: any colorectal dysplasia, high-grade dysplasia and/or colorectal adenocarcinoma (HGD/CRC), and invasive CRC. We validated the method on diagnostic tasks by applying prompts to reports from patients with IBD (and validated separately in non-IBD) and calculated F-1 scores as a balanced accuracy measure. Results In patients with IBD in MVP, we achieved F1-scores of 96.1% (95% CI 92.5-99.4%) for identifying dysplasia, 93.7% (88.2-98.4%) for identifying HGD/CRC, and 98% (96.3-99.4%) for identifying CRC. In patients without IBD in MVP, we achieved F1-scores of 99.2% (98.2-100%) for identifying any colorectal dysplasia, 96.5% (93.0-99.2%) for identifying HGD/CRC, and 95% (92.8-97.2%) for identifying CRC using LLM Gemma-2. Conclusion LLMs provided excellent accuracy in extracting the diagnoses of interest from EHRs. Our validated methods generalized to unstructured pathology notes, even withstanding challenges of resource-limited computing environments. This may therefore be a promising approach for other clinical phenotypes given the minimal human-led development required.
Collapse
Affiliation(s)
- Brian Johnson
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Tyler Bath
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Xinyi Huang
- Veterans Medical Research Foundation, San Diego, CA, USA
| | - Mark Lamm
- Veterans Medical Research Foundation, San Diego, CA, USA
| | - Ashley Earles
- Veterans Medical Research Foundation, San Diego, CA, USA
| | - Hyrum Eddington
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Anna M. Dornisch
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiation Medicine & Applied Sciences, University of California San Diego, La Jolla, CA
| | - Lily J. Jih
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Samir Gupta
- VA San Diego Healthcare System, San Diego, CA, USA
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Shailja C. Shah
- VA San Diego Healthcare System, San Diego, CA, USA
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Kit Curtius
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
28
|
Zhao Z, Xu Y, Hu Y. Acid-resistant chemotactic DNA micromotors for probiotic delivery in inflammatory bowel disease. Nat Commun 2025; 16:3778. [PMID: 40263286 PMCID: PMC12015548 DOI: 10.1038/s41467-025-59172-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 04/13/2025] [Indexed: 04/24/2025] Open
Abstract
Microcapsules composed of synthetic polymeric matrices have attracted considerable attention in delivering oral probiotics. However, existing polymeric microcapsules demonstrate inadequate acid resistance and adaptability, as well as deficiency in the inflamed colon-specificity and uncontrolled release of probiotics therein. Herein, a DNA microcapsule is prepared as a probiotic-transporting micromotor through photo-crosslinking of hyaluronic acid methacrylate and acrydite-modified A-/C-rich oligomers within the microfludically generated droplets in the presence of nitric oxide-cleavable crosslinker and gas donor manganese carbonyl (MnCO). As the microcapsules traverse stomach, duodenum, and ultimately colon, the formation and dissociation of A-motif and i-motif structures instigate a reversible shrinking-swelling transition of microcapsules to preserve probiotic viability. Subsequently, the microcapsules exhibit chemotaxis towards inflamed colon site, driven by a gas-generating reaction between MnCO and elevated reactive oxygen species. Following disintegration of the microcapsules, triggered by endogenous nitric oxide, probiotics are released to reshape the dysbiosis of intestinal microflora. This advanced delivery system offers significant promise for the effective clinical management of inflammatory bowel disease.
Collapse
Affiliation(s)
- Zinan Zhao
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Yao Xu
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Yong Hu
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, 201804, P. R. China.
| |
Collapse
|
29
|
Hu Z, Li W, Wei L, Ma J. Lactoferrin in cancer: Focus on mechanisms and translational medicine. Biochim Biophys Acta Rev Cancer 2025; 1880:189330. [PMID: 40274081 DOI: 10.1016/j.bbcan.2025.189330] [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: 12/05/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Lactoferrin is an iron-binding glycoprotein that provides natural protective effects to the human body. Its biological properties, including antibacterial, antiviral, anti-inflammatory, immune-regulatory, and iron metabolism-regulating functions, have been extensively studied. With further research, lactoferrin's impact on tumorigenesis and tumor microenvironment has become increasingly evident, as it inhibits tumor proliferation, invasion, and metastasis through multiple pathways. This article summarizes the molecular mechanisms underlying lactoferrin's anticancer effects, explores its association with the malignant progression of various cancers, and highlights its clinical translational potential as a potential cancer biomarker and drug delivery carrier to enhance anticancer therapy efficiency. Due to the high safety profile of lactoferrin, its widespread application in the field of cancer treatment is highly anticipated.
Collapse
Affiliation(s)
- Zhengyu Hu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medicine Sciences, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Wenchao Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medicine Sciences, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Lingyu Wei
- Laboratory of Clinical Research Center, Department of Pathology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China.
| | - Jian Ma
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medicine Sciences, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China.
| |
Collapse
|
30
|
Ramineni M, Ettel M, Hao Y, Liao X. SATB2 Loss Is a Sensitive Biomarker for Dysplasia in Inflammatory Bowel Disease. J Transl Med 2025; 105:104179. [PMID: 40258492 DOI: 10.1016/j.labinv.2025.104179] [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: 01/04/2025] [Revised: 03/18/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025] Open
Abstract
Loss of SATB2 expression has emerged as a promising biomarker for dysplasia in inflammatory bowel disease (IBD), but its sensitivity and specificity remain unclear. We retrospectively evaluated immunohistochemical (IHC) staining of SATB2 and p53 in colorectal biopsies from 37 IBD patients (25 men and 12 women; median age: 48 years) with suspected dysplasia. The cohort included 26 ulcerative colitis (70%) and 11 Crohn's disease (30%). Fourteen patients (38%) developed IBD-associated invasive carcinoma, and 18 (49%) had persistent dysplasia on follow-up. Histologic review identified 80 lesions initially diagnosed as negative (16%), indefinite (39%), low-grade (36%), and high-grade (9%) dysplasia. IHC revealed aberrant p53 in 35 lesions (44%) and SATB2 loss in 42 lesions (53%), with 19 (24%) showing both abnormalities. Reappraisal of diagnoses combining histology and IHC reclassified lesions into indefinite (20%), low-grade (63%), and high-grade (17%) dysplasia. Lesions with SATB2 loss alone were more frequently of lower grade (P = .003). Dysplasia types included 15 conventional dysplasia (19%) and 65 nonconventional dysplasia (81%). The rates of p53 abnormality, SATB2 loss, and their combination were similar in nonconventional dysplasia (45%, 55%, and 75%, respectively) and conventional dysplasia (40%, 47%, and 67%, respectively) and comparable between cancer patients (50%, 56%, and 74%, respectively) and noncancer patients (39%, 50%, and 72%, respectively). Missed dysplasias in cancer patients were all nonconventional, and lesions with p53 abnormality more likely progressed to cancer (P = .002). In conclusion, SATB2 loss is a sensitive marker for IBD-associated dysplasia. Combined use of SATB2 and p53 IHC improves dysplasia detection and reduces false-negative diagnosis, supporting its application into routine diagnostic practice.
Collapse
Affiliation(s)
- Madhurya Ramineni
- Department of Pathology, University of Rochester Medical Center, Rochester, New York
| | - Mark Ettel
- Department of Pathology, University of Rochester Medical Center, Rochester, New York
| | - Yansheng Hao
- Department of Pathology, University of Rochester Medical Center, Rochester, New York
| | - Xiaoyan Liao
- Department of Pathology, University of Rochester Medical Center, Rochester, New York.
| |
Collapse
|
31
|
Yuan J, Ma J, Zhang F, Wang T, Jian X, Wang B, Li W, Zhang X, Cao Y, Yang H, Ma Y, Wang H. Neutrophil-derived serine proteases induce FOXA2-mediated autophagy dysfunction and exacerbate colitis-associated carcinogenesis via protease activated receptor 2. Autophagy 2025:1-18. [PMID: 40205686 DOI: 10.1080/15548627.2025.2489335] [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: 09/04/2024] [Revised: 03/22/2025] [Accepted: 04/02/2025] [Indexed: 04/11/2025] Open
Abstract
Autophagy plays a critical role in colitis-associated colorectal cancer (CAC). However, non-autonomous regulation of macroautophagic/autophagic flux during inflammation remains largely unexplored. Here, we show that F2rl1/Par2 deficiency (F2rl1[ΔIEC]) aggravated azoxymethane-dextran sulfate sodium-induced CAC based on tumor number and burden, promoted autophagy dysfunction characterized by SQSTM1/p62 accumulation and autophagosome-lysosome fusion inhibition in IECs, and reduced lysosomal acidification by suppressing FOXA2-induced V-ATPase ATP6V0E1 transcription. FOXA2 or ATP6V0E1 overexpression rescued autophagy impairment, reactive oxygen species accumulation, and DNA damage induced by F2RL1 deficiency in vitro and in vivo. Neutrophil-derived serine proteases suppressed FOXA2 expression, causing autophagy dysfunction. F2RL1 knockout completely blocked the effects of neutrophil proteases on FOXA2 and ATP6V0E1. The correlation between neutrophil and FOXA2-ATP6V0E1 activities was validated in ulcerative colitis and colorectal carcinoma. Therefore, F2RL1 deficiency in intestinal epithelial cells suppressed FOXA2 expression, leading to V-ATPase-mediated autophagic dysfunction and exacerbating CAC. Neutrophils may contribute to impaired autophagy and promote CAC by inactivating canonical F2RL1/PAR2 signaling via its derived proteases. F2RL1/PAR2 signaling may participate in maintaining intestinal homeostasis via autophagy. These findings provide useful insights into F2RL1/PAR2 and its cleaving serine proteases in CAC and would help in developing new therapeutic strategies for this malignancy.Abbreviations: AOM: azoxymethane; ATP6V0C: ATPase H+ transporting V0 subunit c; ATP6V0E1: ATPase H+ transporting V0 subunit e1; ATP6V1C2: ATPase H+ transporting V1 subunit C2; ATP6V1F: ATPase H+ transporting V1 subunit F; CAC: colitis-associated colorectal cancer; CRC: colorectal cancer; CTSB: cathepsin B; CTSG: cathepsin G; DEGs: differentially expressed genes; DSS: dextran sulfate sodium; FOXA2: forkhead box protein A2; F2RL1: F2R like trypsin receptor 1; IBD: inflammatory bowel disease; IECs: intestinal epithelial cells; LAMP1: lysosomal associated membrane protein 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; ROS: reactive oxygen species; SQSTM1/p62: sequestosome 1; TFs: transcription factors; UC: ulcerative colitis.
Collapse
Affiliation(s)
- Junhu Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianhui Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fanyu Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tan Wang
- State Key Lab of Molecular Oncology, Laboratory of Cell and Molecular Biology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaxiang Jian
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Bingzhi Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiwei Li
- Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiaoli Zhang
- Department of Injury and Repair, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yubin Cao
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Hong Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yiming Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
32
|
Yang H, Zhang X, Wu J, Xiao Y, Dai L, Wang G, Zhang X, Hu C, He S, Yuan Z. Probiotic Membrane-Modified Nanocomposite Alleviates Inflammation and Microbiota Dysbiosis in Colitis by Scavenging Oxidative Stress and Restoring Immune Homeostasis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22245-22265. [PMID: 40184333 DOI: 10.1021/acsami.4c22004] [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: 04/06/2025]
Abstract
Inflammatory bowel disease (IBD) is a complex chronic intestinal disorder in which excessive oxidative stress, dysregulated immune response, and microbiota dysbiosis contribute to recurrent episodes of inflammation in the colonic mucosa. Current clinical treatments focusing solely on inflammation resolution often exhibit limited efficacy due to the inability to fundamentally improve the pathological microenvironment. Herein, a probiotic membrane-modified drug delivery nanocomposite, namely, MPDA@Cur@EM, is developed for the comprehensive treatment of IBD. It contains two components: the curcumin-loaded mesoporous polydopamine nanoparticle (MPDA@Cur) as the core and the Escherichia coli Nissle 1917 outer membrane (EM) as the surface. For MPDA@Cur, the pathological microenvironment triggers the responsive release of curcumin. Importantly, MPDA@Cur can effectively alleviate the inflammatory response of LPS-activated macrophages through MPDA-mediated ROS scavenging and curcumin-induced M2 polarization. In the dextran sulfate sodium (DSS)-induced colitis model, the EM coating not only allows for the targeting enrichment of orally administered MPDA@Cur@EM to the inflamed colonic mucosa, but also participates in the regulation of intestinal flora. Consequently, MPDA@Cur@EM efficiently attenuates the inflammatory reaction and restores the intestinal barrier functions, demonstrated by the multipronged manner of restoring redox balance, remodeling immune homeostasis, and reshaping the gut microecology. Collectively, this work provides a safe and promising codelivery strategy of probiotic product, antioxidative nanoenzyme, and therapeutic drug for comprehensive management of IBD.
Collapse
Affiliation(s)
- Huan Yang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Xu Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Jianshuang Wu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Yao Xiao
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Liangliang Dai
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Gaoyang Wang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Xiaohong Zhang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Chenghu Hu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Shuixiang He
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Zhang Yuan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| |
Collapse
|
33
|
Balajthy Z, Szaszák P, Almási S, Lantos T, Sejben A. Evaluation of dysplasias associated with inflammatory bowel disease-a single-center, retrospective, 5-year experience. Pathol Oncol Res 2025; 31:1612105. [PMID: 40303424 PMCID: PMC12037400 DOI: 10.3389/pore.2025.1612105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025]
Abstract
Introduction Several novel morphological variants of inflammatory bowel disease (IBD)- associated dysplasias have been described in recent years. The objective of our study was to reevaluate some of our IBD-associated neoplasia cases and retrospectively identify the so-called non-conventional dysplasias (NCDs). Methods We established a database of IBD patients registered between 2011 and 2015 at the Department of Pathology, University of Szeged. Patients with neoplastic samples were extracted into a separate database. Clinical and pathological characteristics were documented for each case. Histological slides were retrospectively reviewed, and cases were reclassified. Results During the study period, 57 patients had neoplastic samples, and 47 patients were identified with conventional dysplasias (82.5%). A significant association was found between conventional dysplasias and dysplasia localization (P = 0.004), size (P = 0.012), endoscopic appearance (P = 0.006), grade (P = 0.011), macroscopic appearance of colorectal carcinoma (P = 0.009), and pT stage (P = 0.01). NCD was identified in 20 cases (35.1%). The most frequently observed subtype was serrated not otherwise specified (NOS) dysplasia (n = 6; 30%). Significant associations were detected between the development of NCD and several clinical-pathological features, including the occurrence (P < 0.001), localization (P = 0.001), size (P = 0.002), macroscopic appearance (P = 0.01), grade (P = 0.005), histological subtype (P = 0.003), pT (P = 0.003) and pM stage (P = 0.047) of colorectal carcinoma, as well as microsatellite status (P < 0.001). Discussion The identification of IBD-associated NCDs might play a crucial role in future clinical practice. Some authors suggest closer patient follow-up upon identification of these lesions and recommend random biopsy sampling in IBD patients to detect potentially occult lesions. Further studies involving larger national and international patient cohorts are warranted to gain a more comprehensive understanding of the clinical behavior of NCDs.
Collapse
Affiliation(s)
- Zsófia Balajthy
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Panna Szaszák
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Szintia Almási
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Tamás Lantos
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Anita Sejben
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| |
Collapse
|
34
|
Hisamatsu T, Miyoshi J, Oguri N, Morikubo H, Saito D, Hayashi A, Omori T, Matsuura M. Inflammation-Associated Carcinogenesis in Inflammatory Bowel Disease: Clinical Features and Molecular Mechanisms. Cells 2025; 14:567. [PMID: 40277893 PMCID: PMC12025475 DOI: 10.3390/cells14080567] [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: 03/23/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Inflammatory bowel disease (IBD), comprising ulcerative colitis (UC) and Crohn's disease (CD), is a chronic condition marked by persistent intestinal inflammation of unknown etiology. Disease onset involves genetic predisposition and environmental factors that disrupt the intestinal immune homeostasis. The intestinal microbiome and immune response play pivotal roles in disease progression. Advances in molecular therapies and early interventions have reduced surgery rates; however, colorectal cancer (CRC) remains a significant concern, driven by chronic inflammation. In UC, the risk of UC-associated neoplasia (UCAN) increases with disease duration, while CD patients face elevated risks of small intestine, anal fistula, and anal canal cancers. Endoscopic surveillance is advised for UCAN, but optimal screening intervals remain undefined, and no established guidelines exist for CD-associated cancers. UCAN morphology often complicates detection due to its flat, inflammation-blended appearance, which differs pathologically from sporadic CRC (sCRC). UCAN is frequently surrounded by dysplasia, with p53 mutations evident at the dysplasia stage. IBD-associated gastrointestinal cancers exemplify inflammation-driven carcinogenesis with distinct molecular mechanisms from the adenoma-carcinoma sequence. This review explores the epidemiology, risk factors, clinical and pathological features, current surveillance practices, and molecular pathways underlying inflammation-associated cancers in IBD.
Collapse
Affiliation(s)
- Tadakazu Hisamatsu
- Department of Gastroenterology and Hepatology, Kyorin University School of Medicine, Tokyo 181-8611, Japan; (J.M.); (N.O.); (H.M.); (D.S.); (T.O.); (M.M.)
| | - Jun Miyoshi
- Department of Gastroenterology and Hepatology, Kyorin University School of Medicine, Tokyo 181-8611, Japan; (J.M.); (N.O.); (H.M.); (D.S.); (T.O.); (M.M.)
| | - Noriaki Oguri
- Department of Gastroenterology and Hepatology, Kyorin University School of Medicine, Tokyo 181-8611, Japan; (J.M.); (N.O.); (H.M.); (D.S.); (T.O.); (M.M.)
| | - Hiromu Morikubo
- Department of Gastroenterology and Hepatology, Kyorin University School of Medicine, Tokyo 181-8611, Japan; (J.M.); (N.O.); (H.M.); (D.S.); (T.O.); (M.M.)
| | - Daisuke Saito
- Department of Gastroenterology and Hepatology, Kyorin University School of Medicine, Tokyo 181-8611, Japan; (J.M.); (N.O.); (H.M.); (D.S.); (T.O.); (M.M.)
- Department of Gastroenterology and Hepatology, Kyorin University Suginami Hospital, Tokyo 166-0012, Japan
| | - Akimasa Hayashi
- Department of Pathology, Kyorin University School of Medicine, Tokyo181-8611, Japan;
| | - Teppei Omori
- Department of Gastroenterology and Hepatology, Kyorin University School of Medicine, Tokyo 181-8611, Japan; (J.M.); (N.O.); (H.M.); (D.S.); (T.O.); (M.M.)
- Department of Gastroenterology and Hepatology, Kyorin University Suginami Hospital, Tokyo 166-0012, Japan
| | - Minoru Matsuura
- Department of Gastroenterology and Hepatology, Kyorin University School of Medicine, Tokyo 181-8611, Japan; (J.M.); (N.O.); (H.M.); (D.S.); (T.O.); (M.M.)
| |
Collapse
|
35
|
Zheng Z, Jin W, Guo W, Jin Z, Zuo Y. Oral Fusobacterium nucleatum exacerbates ulcerative colitis via the oral-gut axis: mechanisms and therapeutic implications. Front Cell Infect Microbiol 2025; 15:1564169. [PMID: 40260115 PMCID: PMC12009839 DOI: 10.3389/fcimb.2025.1564169] [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: 01/21/2025] [Accepted: 03/18/2025] [Indexed: 04/23/2025] Open
Abstract
Background Fusobacterium nucleatum (F. nucleatum) is an anaerobic bacterium known for its association with periodontal disease and oral infections. It has been implicated in the development of gastrointestinal diseases such as inflammatory bowel disease and colorectal cancer. Ulcerative colitis (UC), which is characterized by chronic inflammation of the colon, is a condition of unknown etiology with a rising incidence rate, significantly affecting the quality of life for patients. The increased intestinal permeability during UC may facilitate the adherence or invasion of F. nucleatum into the damaged intestinal barrier, leading to exacerbated inflammation. Methods This article introduces the concept of the oral-gut axis, reviewing existing literature to analyze the role of F. nucleatum in the pathogenesis of UC and exploring its potential pathogenic mechanisms. It also summarizes the latest advances in treating patients with UC who have F. nucleatum and looks forward to prospective therapeutic strategies and the translational prospects of F. nucleatum within the oral-gut axis. Results F. nucleatum may be a key player in the pathogenesis of UC, likely due to its invasiveness during periods of increased intestinal permeability. The paper also discusses innovative approaches for the prevention and management of UC exacerbated by F. nucleatum, paving the way for more effective treatment of UC. Conclusion The review offers new insights into the complex relationship between the oral microbiome and intestinal diseases, enhancing our understanding of their dynamic interactions. There is a paucity of literature on therapeutic approaches, indicating a need for further clinical research.
Collapse
Affiliation(s)
- Zhaoyu Zheng
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenqin Jin
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weiwei Guo
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhao Jin
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuling Zuo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
36
|
Yuan Q, Shi Y, Wang J, Xie Y, Li X, Zhao J, Jiang Y, Qiao Y, Guo Y, Zhang C, Lu J, Zhao T, Dong Z, Li P, Dong Z, Liu K. p38 mediated ACSL4 phosphorylation drives stress-induced esophageal squamous cell carcinoma growth through Src myristoylation. Nat Commun 2025; 16:3319. [PMID: 40195298 PMCID: PMC11976994 DOI: 10.1038/s41467-025-58342-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/18/2025] [Indexed: 04/09/2025] Open
Abstract
The comprehension of intricate molecular mechanisms underlying how external stimuli promote malignancy is conducive to cancer early prevention. Esophageal squamous cell carcinoma (ESCC) is considered as an external stimuli (hot foods, tobacco, chemo-compounds) induced cancer, characterized by stepwise progression from hyperplasia, dysplasia, carcinoma in situ and invasive carcinoma. However, the underlying molecular mechanism governing the transition from normal epithelium to neoplastic processes in ESCC under persistent external stimuli has remained elusive. Herein, we show that a positive correlation between p38 and ERK1/2 activation during the progression of ESCC. We identify that phosphorylation of ACSL4 at T679 by p38 enhances its enzymatic activity, resulting in increased production of myristoyl-CoA (C14:0 CoA). This subsequently promotes Src myristoylation and activates downstream ERK signaling. Our results partially elucidate the role of ACSL4 in mediating stress-induced signaling pathways that activate growth cascades and contribute to tumorigenesis.
Collapse
Affiliation(s)
- Qiang Yuan
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Yunshu Shi
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Junyong Wang
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
| | - Yifei Xie
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoyu Li
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Jimin Zhao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Yanan Jiang
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Yan Qiao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Yaping Guo
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Chengjuan Zhang
- Center of Bio-Repository, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Lu
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Tongjin Zhao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Zhongshan Hospital, Fudan University, Shanghai Qi Zhi Institute, Shanghai, China
| | - Ziming Dong
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Peng Li
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China.
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China.
| | - Zigang Dong
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China.
| | - Kangdong Liu
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China.
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
37
|
Liang Y, Miao Z, Chen J, Tan L, Zhao Y, Cui X, Zhong J, Zhong R, Yue W, Qiu B, Yu H, He C. Glycine tabacina extract alleviates inflammatory bowel disease via NF-κB, JNK and Nrf2 signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119744. [PMID: 40199409 DOI: 10.1016/j.jep.2025.119744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the colon, often triggered by unhealthy diets, infections, and dysregulated immune responses. Current treatments for IBD are limited by relapse, drug resistance, side effects, and high costs. Glycine tabacina (Labill.) Benth, a legume native to southeastern China, has traditionally been used for its medicinal properties in treating rheumatoid arthritis, nephritis, and osteoporosis. However, its effects on IBD remain unexplored. AIM OF THE STUDY This study aimed to investigate the anti-colitis effects and underlying mechanisms of Glycine tabacina ethanol extract (GTE) using in vitro and in vivo models. MATERIALS AND METHODS The chemical components of GTE were identified using high-performance liquid chromatography (HPLC). The effects of GTE on lipopolysaccharide (LPS)-induced inflammation and oxidative stress were assessed in Caco-2 cells. Dextran sulfate sodium (DSS)-induced colitis in mice was used to evaluate GTE's therapeutic potential. ELISA, RT-qPCR, immunofluorescence, and immunoblotting were performed to measure gene expression and signaling pathway activity. Histological analysis of colon tissues was conducted using H&E staining. RESULTS GTE significantly reduced LPS-induced inflammation and oxidative stress in Caco-2 cells and alleviated DSS-induced colitis in mice. Mechanistically, GTE decreased pro-inflammatory cytokines, matrix metalloproteinases (MMPs), and reactive oxygen species (ROS), while improving intestinal barrier integrity. Furthermore, GTE suppressed the NF-κB and MAPK/JNK pathways while activating the Nrf2 pathway. These results suggest that GTE may serve as a promising therapeutic agent for IBD by modulating key inflammatory and oxidative stress pathways. CONCLUSIONS The anti-inflammatory and antioxidant properties of GTE mitigated intestinal epithelial cell damage by preserving tight junction proteins and maintaining intestinal barrier integrity. Given its high efficacy and favorable safety profile, GTE represents a promising therapeutic candidate for managing chronic and refractory inflammatory disorders such as IBD.
Collapse
Affiliation(s)
- Yongkai Liang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Zhimin Miao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Junming Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Lihua Tan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Yuxin Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Xiaobing Cui
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Jinmiao Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Ruting Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Wendi Yue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Boyang Qiu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Hua Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, 999078, China; Department of Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, 999078, China; Department of Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China.
| |
Collapse
|
38
|
Pedersen TA, Engjom T, Dimcevski GG, Botteri E, Seip B, Havre RF. Differences in colonoscopy performance among four endoscopy centers in Western Norway: Influence of case-mix. Endosc Int Open 2025; 13:a25469515. [PMID: 40230559 PMCID: PMC11996018 DOI: 10.1055/a-2546-9515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/14/2025] [Indexed: 04/16/2025] Open
Abstract
Background and study aims Unmodifiable patient factors such as age, sex, and indication (case-mix) may influence colonoscopy performance. In this study, we explored how case-mix affected polyp detection, cecal intubation, and pain on a center level. Methods A cross-sectional study was performed on data from four centers in Western Norway registered in the national endoscopy quality registry, Gastronet, in 2020 and 2021. We extracted demographics, indication, and the performance measures cecal intubation rate (CIR), proportion of at least one polyp ≥ 5 mm in size per colonoscopy (PDR-5), and pain. We also analyzed the explanatory variables bowel preparation, withdrawal time, and sedation/analgesia. Results First colonoscopies in 14,765 patients were included. Median age was 60 years (interquartile range 46-71) and 54% were women. Case-mix differed between centers and significantly influenced performance measures. Increased PDR-5 was associated with higher age and male sex (odds ratio [OR] 1.27, 95% confidence interval [CI] 1.18-1.37). The indication surveillance had the highest PDR-5 (44.9%, 95% CI 42.6-47.1) and inflammatory bowel disease the lowest (14.6%, 95% CI 12.3-16.8). CIR decreased with increasing age. Men had less pain (OR 0.33, 95% CI 0.27-0.39). Among indications, surveillance and IBD had higher CIRs and less pain. Performance measures differed among centers, even after adjustment for case-mix and other known explanatory variables such as sedation/analgesia and bowel preparation. Conclusions Case-mix influenced performance measures. Although we showed center differences in performance, other factors, such as individual endoscopist skills, probably influence performance measures. Our study demonstrates the importance of considering case-mix when assessing colonoscopy performance.
Collapse
Affiliation(s)
- Tom Andre Pedersen
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
- Bergen Research group for Advanced Gastrointestinal Endoscopy (BRAGE), Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Trond Engjom
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Georg Gjorgji Dimcevski
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Private outpatient endoscopy centre, Kanalspesialistene AS, Bergen, Norway
| | - Edoardo Botteri
- Section for Colorectal Cancer screening, Cancer Registry of Norway, Oslo, Norway
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Birgitte Seip
- Department of Medicine, Vestfold Hospital Trust, Tonsberg, Norway
| | - Roald Flesland Havre
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Bergen Research group for Advanced Gastrointestinal Endoscopy (BRAGE), Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
39
|
Silvestro O, Vicario CM, Costa L, Sparacino G, Lund-Jacobsen T, Spatola CAM, Merlo EM, Viola A, Giorgianni CM, Catalano A, Fries W, Lo Coco G, Martino G. Defense mechanisms and inflammatory bowel diseases: a narrative review. RESEARCH IN PSYCHOTHERAPY (MILANO) 2025. [PMID: 40178111 DOI: 10.4081/ripppo.2025.854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Growing evidence highlights the crucial role of defense mechanisms in the context of chronic diseases. However, few studies have evaluated the impact of these implicit emotion regulation strategies on the adaptation processes related to inflammatory bowel diseases (IBD). This narrative review aimed to explore the role of defense mechanisms in patients with IBD and clarify their association with related psychological and physical symptoms. A literature search was conducted using PubMed and PsycINFO databases to select studies considering defense mechanisms in patients with IBD. Inclusion criteria were English language articles, diagnosis of Crohn's disease or ulcerative colitis, and use of validated assessment instruments specifically related to defense mechanisms. Six studies, including a total of 664 patients, were deemed eligible. Immature defense mechanisms were commonly detected in IBD patients, with significant effects on psychological and physical health. Significant associations were found between defense mechanisms, perceived health-related quality of life (HR-QoL), and psychological distress. Findings suggested that immature defense mechanisms may negatively impact the management of disease, leading to lower perceived HR-QoL, decreased treatment adherence, and increased risk of psychopathological symptoms. Considering these findings, we suggest that an integrated clinical evaluation, including an in-depth investigation of defense mechanisms, may promote more effective psychological treatments and improve psychological well-being in patients suffering from IBD.
Collapse
Affiliation(s)
- Orlando Silvestro
- Department of Health Sciences, University Magna Graecia of Catanzaro
| | - Carmelo M Vicario
- Department of Cognitive Science, Psychology, Education and Cultural Studies, University of Messina
| | - Ludovico Costa
- Course Degree in Clinical and Health Psychology in the Life Cycle, University of Messina
| | | | - Trine Lund-Jacobsen
- Department of Endocrinology, Centre for Cancer and Organ Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen
| | | | - Emanuele M Merlo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina
| | - Anna Viola
- Department of Clinical and Experimental Medicine, University of Messina
| | - Concetto M Giorgianni
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina
| | - Antonino Catalano
- Department of Clinical and Experimental Medicine, University of Messina
| | - Walter Fries
- Department of Clinical and Experimental Medicine, University of Messina
| | - Gianluca Lo Coco
- Department of Psychology, Educational Science and Human Movement, University of Palermo
| | - Gabriella Martino
- Department of Clinical and Experimental Medicine, University of Messina
| |
Collapse
|
40
|
Zhu Y, Cao S. Unraveling the Complexities of Myeloid-Derived Suppressor Cells in Inflammatory Bowel Disease. Int J Mol Sci 2025; 26:3291. [PMID: 40244120 PMCID: PMC11989781 DOI: 10.3390/ijms26073291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) regulate immune responses in many pathological conditions, one of which is inflammatory bowel disease (IBD), an incurable chronic disorder of the digestive tract and beyond. The pathophysiology of IBD remains unclear, likely involving aberrant innate and adaptive immunity. Studies have reported altered population of MDSCs in patients with IBD. However, their distribution varies among patients and different preclinical models of IBD. The expansion and activation of MDSCs are likely driven by various stimuli during intestinal inflammation, but the in-depth mechanisms remain poorly understood. The role of MDSCs in the pathogenesis of IBD appears to be paradoxical. In addition to intestinal inflammation, suppressive MDSCs may promote colitis-to-colon cancer transition. In this Review, we summarize recent progresses on the features, activation, and roles of MDSCs in the development of IBD and IBD-associated colon cancer.
Collapse
Affiliation(s)
| | - Siyan Cao
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
| |
Collapse
|
41
|
Li T, Wu X, Li X, Chen M. Cancer-associated fungi: An emerging powerful player in cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189287. [PMID: 39971202 DOI: 10.1016/j.bbcan.2025.189287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
The role of the human microbiome in cancer has been extensively studied, focusing mainly on bacteria-host interactions and their impact on tumor development and treatment response. However, fungi, an immune-active component of the human microbiome, have received less attention regarding their roles in cancer. Recent studies have identified the widespread and specific colonization and distribution of fungi in multiple sites in patients across various cancer types. Importantly, host-fungal immune interactions significantly influence immune regulation within the tumor microenvironment. The rapid advancement of immune-checkpoint blockade (ICB)-based cancer immunotherapy creates an urgent need for effective biomarkers and synergistic therapeutic targets. Cancer-associated fungi and their associated antifungal immunity demonstrate significant potential and efficacy in enhancing cancer immunotherapy. This review summarizes and discusses the growing evidence of the functions and mechanisms of commensal and pathogenic cancer-associated fungi in cancer immunotherapy. Additionally, we emphasize the potential of fungi as predictive biomarkers and therapeutic targets in cancer immunotherapy.
Collapse
Affiliation(s)
- Tianhang Li
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China; Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.
| | - Xiangyu Wu
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiangyang Li
- Department of Gastrointestinal Tumor Surgery, Nanjing Tianyinshan Hospital, Affiliated Hospital of China Pharmaceutical University, Nanjing, China.
| | - Ming Chen
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China; Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.
| |
Collapse
|
42
|
Huang B, An H, Gui M, Qiu Y, Xu W, Chen L, Li Q, Yao S, Lin S, Khrustaleva TA, Wang R, Lin J. Qingjie Fuzheng Granule prevents colitis-associated colorectal cancer by inhibiting abnormal activation of NOD2/NF-κB signaling pathway mediated by gut microbiota disorder. CHINESE HERBAL MEDICINES 2025. [DOI: 10.1016/j.chmed.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025] Open
|
43
|
Itzkowitz SH, Jiang Y, Villagra C, Colombel JF, Sultan K, Lukin DJ, Faleck DM, Scherl E, Chang S, Chen L, Katz S, Kwah J, Swaminath A, Petralia F, Sharpless V, Sachar D, Jandorf L, Axelrad JE. Safety of Immunosuppression in a Prospective Cohort of Inflammatory Bowel Disease Patients With a HIstoRy of CancEr: SAPPHIRE Registry. Clin Gastroenterol Hepatol 2025; 23:855-865.e5. [PMID: 38768673 DOI: 10.1016/j.cgh.2024.05.006] [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: 01/29/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND & AIMS In patients with inflammatory bowel disease (IBD) and a history of cancer, retrospective studies have suggested that exposure to immunosuppressive agents does not increase the risk of incident (recurrent or new) cancer compared with unexposed patients. Safety of Immunosuppression in a Prospective Cohort of Inflammatory Bowel Disease Patients and a HIstoRy of CancEr (SAPPHIRE) is a prospective registry aimed at addressing this issue. METHODS Since 2016, patients with IBD and confirmed index cancer before enrollment were followed up annually. Patients receiving chemotherapy or radiation at enrollment, or recurrent cancer within 5 years, were excluded. The primary outcome was development of incident cancer related to exposure to immunosuppressive medications. RESULTS Among 305 patients (47% male, 88% white), the median age at IBD diagnosis and cancer were 32 and 52 years, respectively. Index cancers were solid organ (46%), dermatologic (32%), gastrointestinal (13%), and hematologic (9%). During a median follow-up period of 4.8 years, 210 patients (69%) were exposed to immunosuppressive therapy and 46 patients (15%) developed incident cancers (25 new, 21 recurrent). In unadjusted analysis, the crude rate of incident cancer in unexposed patients was 2.58 per 100 person-years vs 4.78 per 100 person-years (relative risk, 1.85; 95% CI, 0.92-3.73) for immunosuppression-exposed patients. In a time-varying proportional hazards model adjusting for sex, smoking history, age and stage at index malignancy, and nonmelanoma skin cancer, no significant association was found between receipt of immunosuppression and incident cancer (adjusted hazard ratio, 1.41; 95% CI, 0.69-2.90), or with any major drug class. CONCLUSIONS In this interim analysis of patients with IBD and a history of cancer, despite numerically increased adjusted hazard ratios, we did not find a statistically significant association between subsequent exposure to immunosuppressive therapies and development of incident cancers.
Collapse
Affiliation(s)
- Steven H Itzkowitz
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Yue Jiang
- Department of Statistical Science, Duke University, Durham, North Carolina
| | - Cristina Villagra
- Department of Population Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Keith Sultan
- Division of Gastroenterology, Department of Medicine, Northwell Health-North Shore University Hospital, Zucker School of Medicine, Manhasset, New York
| | - Dana J Lukin
- Division of Gastroenterology, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - David M Faleck
- Gastroenterology, Hepatology and Nutrition Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ellen Scherl
- Division of Gastroenterology, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Shannon Chang
- Division of Gastroenterology, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - LeaAnn Chen
- Division of Gastroenterology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Seymour Katz
- Division of Gastroenterology, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Joann Kwah
- Division of Gastroenterology, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Arun Swaminath
- Division of Gastroenterology, Department of Medicine, Northwell Health-Lenox Hill Hospital, Zucker School of Medicine, New York, New York
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Virginia Sharpless
- Collaborative Studies Coordinating Center, University of North Carolina, Chapel Hill, North Carolina
| | - David Sachar
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lina Jandorf
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Population Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jordan E Axelrad
- Division of Gastroenterology, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| |
Collapse
|
44
|
Nishio M, Hirasawa K, Saigusa Y, Shiqi Z, Kenemura T, Hama T, Atsusaka R, Azuma D, Ozeki Y, Sawada A, Ikeda R, Fukuchi T, Kobayashi R, Sato C, Ogashiwa T, Chiba S, Inayama Y, Kunisaki R, Maeda S. Usefulness of Magnifying Endoscopy With Narrow-Band Imaging for Diagnosis of Ulcerative Colitis-Associated Neoplasia. J Gastroenterol Hepatol 2025; 40:900-906. [PMID: 39763012 DOI: 10.1111/jgh.16877] [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: 07/12/2024] [Revised: 11/28/2024] [Accepted: 12/23/2024] [Indexed: 04/05/2025]
Abstract
BACKGROUND AND AIM Qualitative diagnosis of ulcerative colitis-associated neoplasia (UCAN) is crucial for surveillance colonoscopy in patients with ulcerative colitis (UC). Although the utility of magnifying endoscopy with narrow-band imaging (ME-NBI) in sporadic neoplasia diagnosis has been reported, its efficacy in UCAN remains unclear. This study aimed to evaluate the usefulness of ME-NBI for qualitative diagnosis of UCAN. METHODS We generated 60 ME-NBI images (30 UCANs and 30 nonneoplasia lesions, including 10 polypoid and 20 nonpolypoid lesions) from patients with UC who underwent colonoscopy at our hospital between 2015 and 2023. Eleven endoscopists (seven experts and four trainees) independently assessed these images. Lesions were categorized into high- (≥ 80%), moderate- (50%-79%), and low- (< 50%) accuracy groups on the basis of the correct diagnostic rate. RESULTS Overall sensitivity, specificity, and correct diagnostic rates were 66.5%, 79.0%, and 71.8%, respectively. Experts tended to exhibit higher specificity than trainees (83% vs. 70%). Polypoid lesions showed higher sensitivity (92% vs. 54%) and lower specificity (61% vs. 88%) than nonpolypoid lesions. Overall, the kappa value was 0.411. In UCAN, 37%, 37%, and 24% were classified into the high-, moderate-, and low-accuracy groups, respectively. All endoscopists assessed one case of UCAN in the low-accuracy group as a nonneoplastic vessel with a surface pattern. Only two nonneoplasias were identified as having nonneoplastic vessel and surface patterns by all endoscopists. CONCLUSIONS This study demonstrated the usefulness of ME-NBI for qualitative diagnosis, along with its limitations. A unique endoscopic diagnostic algorithm for UCAN, incorporating ME-NBI and other modalities, is necessary.
Collapse
Affiliation(s)
- Masafumi Nishio
- Division of Endoscopy, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
- Inflammatory Bowel Disease Center, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Kingo Hirasawa
- Division of Endoscopy, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Yusuke Saigusa
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Zhao Shiqi
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Tokomi Kenemura
- Division of Endoscopy, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Takanori Hama
- Division of Endoscopy, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Reo Atsusaka
- Division of Endoscopy, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Daisuke Azuma
- Division of Endoscopy, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Yuichiro Ozeki
- Division of Endoscopy, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Atsushi Sawada
- Division of Endoscopy, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Ryosuke Ikeda
- Division of Endoscopy, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Takehide Fukuchi
- Division of Endoscopy, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Ryosuke Kobayashi
- Division of Endoscopy, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Chiko Sato
- Division of Endoscopy, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Tsuyoshi Ogashiwa
- Inflammatory Bowel Disease Center, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Sawako Chiba
- Division of Diagnostic Pathology, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Yoshiaki Inayama
- Division of Diagnostic Pathology, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Reiko Kunisaki
- Inflammatory Bowel Disease Center, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Shin Maeda
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
45
|
Xu S, Zhang Y, Ding X, Yang Y, Gao J, Zou N, Lu L, He J. Intestinal microbiota affects the progression of colorectal cancer by participating in the host intestinal arginine catabolism. Cell Rep 2025; 44:115370. [PMID: 40022728 DOI: 10.1016/j.celrep.2025.115370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/11/2024] [Accepted: 02/07/2025] [Indexed: 03/04/2025] Open
Abstract
Arginine plays a critical role in colorectal cancer (CRC) progression. We find that arginine catabolism is reduced in the intestinal microbiota of patients with CRC but increased in tumor tissue. We further verify that Escherichia coli can consume arginine via the arginine succinyltransferase (AST) pathway, and gavaging mice with the AST-deficient E. coli Nissle 1917 (ΔacEcN) can inhibit arginine catabolism of the intestinal microbiota, thereby increasing the arginine concentration in the colon. In the azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced CRC mouse model, reduced arginine catabolism in the intestinal microbiota increases the arginine concentration in the tumor microenvironment, thereby activating the nitric oxide (NO) synthesis pathway and polyamine synthesis pathway in tumor tissues, stimulating angiogenesis in the tumor microenvironment, inducing M2 macrophage polarization, and activating the Wingless/Integrated (Wnt)/β-catenin pathway, ultimately accelerating CRC progression. This study reveals that intestinal microbiota can affect CRC progression through arginine catabolism, providing a potential target for the prevention and therapy of CRC.
Collapse
Affiliation(s)
- Siyang Xu
- National Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Yuling Zhang
- National Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Xiaoqi Ding
- National Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Yijun Yang
- National Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Jinge Gao
- National Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Ning Zou
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430079, P.R. China
| | - Li Lu
- Department of Gastrointestinal Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430079, P.R. China.
| | - Jin He
- National Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, P.R. China.
| |
Collapse
|
46
|
Chen C, Sun B, Chen K, Bao H, Tao Y, Zhou J, Yuan X, He L, Lu Z, Chen K, Li Y, Yu C, Chen Y, Zhang Y. Atractylenolide-I restore intestinal barrier function by targeting the S100A9/AMPK/mTOR signaling pathway. Front Pharmacol 2025; 16:1530109. [PMID: 40196359 PMCID: PMC11973269 DOI: 10.3389/fphar.2025.1530109] [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: 04/09/2025] Open
Abstract
Impaired intestinal epithelial barrier function is closely associated with the pathogenesis of ulcerative colitis (UC). Atractylenolide-I (AT-I), a major sesquiterpene derived from the herb Atractylodes macrocephala Koidz., has been reported to alleviate DSS-induced colitis in mice. This study aims to investigated the protective effects of AT-1 on intestinal epithelial barrier function and elucidate it's underlying mechanisms. In vivo, an acute colitis model was established in mice, and transcriptomic analysis to identify differentially expressed genes. In vitro, overexpression plasmids and recombinant protein were used to evaluate their effects on intestinal barrier function, and further analysis of its potential mechanisms.The study found that AT-1 ameliorate DSS-induced acute ulcerative colitis, exhibiting protective effects on the intestinal barrier. Transcriptomic analysis revealed that AT-1 significantly modulated the expression of S100A8 and S100A9. Further investigations indicated that S100A9, rather than S100A8, mediated the expression of tight junction proteins, meanwhile, AT-1 reduces neutrophil activation and subsequent release of S100A9. Mechanistically, recombinant human S100A9 protein was found to induce a decrease in intracellular Ca2+ concentration, while AT-1 regulated the expression of tight junction proteins via modulation of the AMPK/mTOR signaling pathway. AT-1 enhances the recovery of DSS-induced intestinal barrier dysfunction by regulating the recombinant human S100A9 protein-mediated AMPK/mTOR signaling pathway. This study provides new insights into the pathogenesis of ulcerative colitis and suggests potential therapeutic strategies for its treatment.
Collapse
Affiliation(s)
- Chen Chen
- Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Bingjie Sun
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Keming Chen
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Han Bao
- Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Xuzhou City Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Xuzhou, China
| | - Yu Tao
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinyong Zhou
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaomin Yuan
- Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Linhai He
- Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhihua Lu
- Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Kaidi Chen
- Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Li
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chengli Yu
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Yugen Chen
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yinan Zhang
- Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
47
|
Alzahrani AJ, Al-Hebshi BM, Yahia ZA, Al-Judaibi EA, Alsaadi KH, Al-Judaibi AA. Impact of Microbiota Diversity on Inflammatory Bowel Disease. Microorganisms 2025; 13:710. [PMID: 40284547 PMCID: PMC12029714 DOI: 10.3390/microorganisms13040710] [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: 02/07/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic condition that includes two main types, Crohn's disease (CD) and ulcerative colitis (UC), involving inflammation of the gastrointestinal (GI) tract. The exact cause of IBD is unknown but could be a combination of genetic, environmental, and immune system factors. This study investigated the impact of IBD on microbiota diversity by evaluating the differences in microbial composition and the microbiota of a control group (A) of healthy individuals and a group (B) of IBD patients. Sixty biopsies were collected from participants recruited from hospitals in Makkah, Saudi Arabia. Biopsy specimens were taken during colonoscopy examination, and bacterial identification was performed by extracting ribosomal DNA from sigmoid colon biopsies using a DNeasy Blood & Tissue Kit. Metagenomics and bioinformatics analyses were then conducted to analyze and compare the microbiota in the two groups. The results showed that the varieties of core microbiome species were 3.81% greater in the IBD patients than in the members of the control group. Furthermore, the differences between the groups were significantly greater than the variations within each group. Differences between the two groups were detected in the relative abundance of Clostridium nexile, Ruminococcus gnavus, Ruminococcus faecis, and Escherichia coli. These results indicate that microbiota could play a role in the pathogenesis of IBD and suggest that microbial diversity can serve as a biomarker for diagnosing the disease and monitoring its progression.
Collapse
Affiliation(s)
- Ashwag J. Alzahrani
- Department of Biological Sciences, Microbiology Section, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia; (A.J.A.); (B.M.A.-H.); (E.A.A.-J.); (K.H.A.)
| | - Basma M. Al-Hebshi
- Department of Biological Sciences, Microbiology Section, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia; (A.J.A.); (B.M.A.-H.); (E.A.A.-J.); (K.H.A.)
| | - Zolfekar A. Yahia
- Department of Internal Medicine, Al Noor Specialist Hospital, Ministry of Health, Makkah 24242, Saudi Arabia;
| | - Effat A. Al-Judaibi
- Department of Biological Sciences, Microbiology Section, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia; (A.J.A.); (B.M.A.-H.); (E.A.A.-J.); (K.H.A.)
| | - Khloud H. Alsaadi
- Department of Biological Sciences, Microbiology Section, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia; (A.J.A.); (B.M.A.-H.); (E.A.A.-J.); (K.H.A.)
| | - Awatif A. Al-Judaibi
- Department of Biological Sciences, Microbiology Section, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia; (A.J.A.); (B.M.A.-H.); (E.A.A.-J.); (K.H.A.)
| |
Collapse
|
48
|
Damiano OM, Stevens AJ, Kenwright DN, Seddon AR. Chronic Inflammation to Cancer: The Impact of Oxidative Stress on DNA Methylation. FRONT BIOSCI-LANDMRK 2025; 30:26142. [PMID: 40152377 DOI: 10.31083/fbl26142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/10/2024] [Accepted: 11/21/2024] [Indexed: 03/29/2025]
Abstract
The genomic landscape of cancer cells is complex and heterogeneous, with aberrant DNA methylation being a common observation. Growing evidence indicates that oxidants produced from immune cells may interact with epigenetic processes, and this may represent a mechanism for the initiation of altered epigenetic patterns observed in both precancerous and cancerous cells. Around 20% of cancers are linked to chronic inflammatory conditions, yet the precise mechanisms connecting inflammation with cancer progression remain unclear. During chronic inflammation, immune cells release oxidants in response to stimuli, which, in high concentrations, can cause cytotoxic effects. Oxidants are known to damage DNA and proteins and disrupt normal signalling pathways, potentially initiating a sequence of events that drives carcinogenesis. While research on the impact of immune cell-derived oxidants on DNA methylation remains limited, this mechanism may represent a crucial link between chronic inflammation and cancer development. This review examines current evidence on inflammation-associated DNA methylation changes in cancers related to chronic inflammation.
Collapse
Affiliation(s)
- Olivia M Damiano
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Aaron J Stevens
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Diane N Kenwright
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Annika R Seddon
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, 8011 Christchurch, New Zealand
| |
Collapse
|
49
|
Fumarola S, Cianfruglia L, Cecati M, Giammarchi C, Vaiasicca S, Gasparrini M. Polyphenol Intake in Elderly Patients: A Novel Approach to Counteract Colorectal Cancer Risk? Int J Mol Sci 2025; 26:2497. [PMID: 40141143 PMCID: PMC11942013 DOI: 10.3390/ijms26062497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Colorectal cancer (CRC) accounts for approximately 10% of all cancers worldwide with an incidence of approximately 60% in patients older than 70 years. In the elderly, the definition of a better therapeutic strategy depends on several factors including the patient's frailty and comorbidity status, life expectancy, and chemotherapy tolerance. In older patients, adverse drug reactions require a reduction in the dose of treatment, resulting in worse oncologic outcomes. In recent years, an increasing number of studies have focused on the potential effects of polyphenols on human health and their use in cancer therapy. In this comprehensive review, we searched the major databases and summarized experimental data of the most important polyphenols in the CRC chemoprevention, with a focus on the molecular mechanisms involved and the antitumor effects in the elderly population. In vitro and in vivo studies have shown that polyphenols exert chemopreventive activity by modulating cell signaling, resulting in the inhibition of cancer development or progression. However, the efficacy seen in experimental studies has not been confirmed in clinical trials, mainly due to their low bioavailability and non-toxic doses. Further research is needed to increase polyphenol bioavailability and reduce side effects in order to suggest their possible use to increase the efficacy of chemotherapeutic treatment.
Collapse
Affiliation(s)
- Stefania Fumarola
- Advanced Technology Center for Aging Research, Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS-INRCA), 60121 Ancona, Italy; (S.F.); (L.C.)
| | - Laura Cianfruglia
- Advanced Technology Center for Aging Research, Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS-INRCA), 60121 Ancona, Italy; (S.F.); (L.C.)
| | - Monia Cecati
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Cinzia Giammarchi
- Scientific Direction, Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS-INRCA), 60121 Ancona, Italy;
| | - Salvatore Vaiasicca
- Center for Neurobiology of Aging, Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS-INRCA), 60121 Ancona, Italy
| | - Massimiliano Gasparrini
- Department of Agriculture, Food and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| |
Collapse
|
50
|
Ma J, Chen C, Wang N, Fang T, Liu Y, He P, Dong W. Identification of Senescence-Related Genes for the Prediction of Ulcerative Colitis Based on Interpretable Machine Learning Models. J Inflamm Res 2025; 18:3431-3447. [PMID: 40093957 PMCID: PMC11908404 DOI: 10.2147/jir.s508396] [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: 12/10/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
Background Cellular senescence, a hallmark of aging, significantly contributes to the pathology of ulcerative colitis (UC). Despite this, the role of senescence-related genes in UC remains largely undefined. This study seeks to clarify the impact of cellular senescence on UC by identifying key senescence-related genes and developing diagnostic models with potential clinical utility. Methods Clinical data and gene expression profiles were obtained from the Gene Expression Omnibus (GEO) database. Senescence-related differentially expressed genes (sene-DEGs) between patients with UC and healthy controls were identified using various bioinformatics techniques. Functional enrichment and immune infiltration analyses were performed to understand subtype characteristics derived from sene-DEGs through consensus clustering. Machine learning algorithms were employed to select feature genes from sene-DEGs, and their expression was validated across multiple independent datasets and human specimens. A nomogram incorporating these feature genes was created and assessed, with its diagnostic performance evaluated using receiver operating characteristic (ROC) analysis on independent datasets. Results Fourteen senescence-related differential genes were identified between patients with UC and healthy controls. These genes enabled the classification of patients with UC into molecular subtypes via unsupervised clustering. ABCB1 and LCN2 emerged as central hub genes through machine learning and feature importance analysis. ROC analysis verified their diagnostic value across various datasets. Validation in independent datasets and human specimens supported the bioinformatics findings. Furthermore, the expression levels of ABCB1 and LCN2 showed significant associations with immune cell profiles. The logistic regression (LR) model based on these genes demonstrated accurate UC prediction, as confirmed by ROC curve analysis. The nomogram model, constructed with feature genes, exhibited outstanding prediction capabilities, supported by DCA, C index, and calibration curve assessments. Conclusion This integrated bioinformatics approach identified ABCB1 and LCN2 as significant biomarkers associated with cellular senescence. These findings enhance the understanding of cellular senescence in UC pathogenesis and propose its potential as a valuable diagnostic biomarker.
Collapse
Affiliation(s)
- Jingjing Ma
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Chen Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Nian Wang
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Ting Fang
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Yinghui Liu
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Pengzhan He
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| |
Collapse
|