1
|
Birgersson M, Holm M, Gallardo-Dodd CJ, Chen B, Stepanauskaitė L, Hases L, Kutter C, Archer A, Williams C. Intestinal estrogen receptor beta modulates the murine colon tumor immune microenvironment. Cancer Lett 2025; 622:217661. [PMID: 40120798 DOI: 10.1016/j.canlet.2025.217661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Chronic inflammation contributes to the development of colorectal cancer, partly through its regulation of the microenvironment and antitumor immunity. Interestingly, women have a lower incidence of colorectal cancer, and estrogen treatment has been shown to reduce the occurrence of colorectal tumors. While intestinal estrogen receptor beta (ERβ, Esr2) can protect against colitis and colitis-induced cancer in mice, its role in shaping the tumor microenvironment remains unknown. In this study, we performed RNA sequencing to analyze the transcriptome of colonic epithelia and tumors from azoxymethane/dextran sulfate sodium-treated wild-type and intestinal ERβ knockout (ERβKOVil) mice and vehicle-treated controls. This revealed significant differences in gene expression and enriched biological processes influenced by sex and genotype, with immune-related responses being overrepresented. Deconvolution supported differential immune cell abundance and immunostaining showed that tumors from ERβKOVil mice displayed significantly increased macrophage infiltration, decreased T cell infiltration, and impaired natural killer cell infiltration. Further, ERβ mRNA levels in clinical colorectal tumors correlated with immune signaling profiles and better survival. Our findings indicate that intestinal ERβ promotes an antitumor microenvironment and could potentially affect the effectiveness of immunotherapy. These insights highlight the importance of ERβ in modulating antitumor immunity and underscore its therapeutic potential in colorectal cancer.
Collapse
Affiliation(s)
- Madeleine Birgersson
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Matilda Holm
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Carlos J Gallardo-Dodd
- Department of Microbiology, Tumor and Cell Biology, SciLifeLab, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Baizhen Chen
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden
| | - Lina Stepanauskaitė
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Linnea Hases
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Claudia Kutter
- Department of Microbiology, Tumor and Cell Biology, SciLifeLab, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Amena Archer
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Cecilia Williams
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 21 Solna, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 83, Huddinge, Sweden.
| |
Collapse
|
2
|
Xu DQ, Geng JX, Gao ZK, Fan CY, Zhang BW, Han X, He LQ, Dai L, Gao S, Yang Z, Zhang Y, Arshad M, Fu Y, Mu XQ. To explore the potential combined treatment strategy for colorectal cancer: Inhibition of cancer stem cells and enhancement of intestinal immune microenvironment. Eur J Pharmacol 2025; 998:177533. [PMID: 40120791 DOI: 10.1016/j.ejphar.2025.177533] [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: 10/05/2024] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND The antibiotic salinomycin, a well-known cancer stem cell inhibitor, may impact the diversity of the intestinal microbiota in colorectal cancer (CRC) mice, which plays a pivotal role in shaping the immune system. This study explores the anti-cancer effects and mechanisms of combining salinomycin and fecal microbiota transplantation (FMT) in treating CRC. METHODS FMT was given via enema, while salinomycin was injected intraperitoneally into the CRC mouse model induced by azoxymethane/dextran sodium sulfate. RESULTS In CRC mice, a large number of LGR5-labeled cancer stem cells and severe disturbances in the intestinal microbiota were observed. Interestingly, salinomycin inhibited the proliferation of cancer stem cells without exacerbating the microbial disorder as expected. In comparison to salinomycin treatment, the combination of salinomycin and FMT significantly improved pathological damage and restored intestinal microbial diversity, which is responsible for shaping the anti-cancer immune microenvironment. The supplementation of FMT significantly increased the levels of propionic acid and butyric acid while also promoting the infiltration of CD8+ T cells and Ly6G+ neutrophils, as well as reducing F4/80+ macrophage recruitment. Notably, cytokines that were not impacted by salinomycin exhibited robust reactions to alterations in the gut microbiota. These included pro-inflammatory factors (IL6, IL12b, IL17, and IL22), chemokine-like protein OPN, and immunosuppressive factor PD-L1. CONCLUSIONS Salinomycin plays the role of "eliminating pathogenic qi," targeting cancer stem cells; FMT plays the role of "strengthening vital qi," reversing the intestinal microbiota disorder and enhancing anti-cancer immunity. They have a synergistic effect on the development of CRC.
Collapse
Affiliation(s)
- Dan-Qi Xu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Jia-Xin Geng
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Zhan-Kui Gao
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Chao-Yuan Fan
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Bo-Wen Zhang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Xing Han
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Li-Qian He
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Lin Dai
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Shuo Gao
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Zhou Yang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Yang Zhang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Muhammad Arshad
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Yin Fu
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, 150006, China.
| | - Xiao-Qin Mu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
3
|
Liu M, Yang C, Peng X, Zheng S, He H, Wang W, Li Y. Formononetin suppresses colitis-associated colon cancer by targeting lipid synthesis and mTORC2/Akt signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156665. [PMID: 40318528 DOI: 10.1016/j.phymed.2025.156665] [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: 12/04/2024] [Revised: 03/08/2025] [Accepted: 03/18/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Colitis-associated colon cancer (CAC) is a serious gastrointestinal malignancy, with a significantly increased incidence among patients with inflammatory bowel disease, posing major challenges to patients' quality of life and prognosis. Modern research highlights Formononetin (FN) for its significant anti-inflammatory and extensively studied anti-cancer properties; however, its precise mechanisms, particularly in CAC, remain unclear and warrant further investigation. OBJECTIVE To investigate the anti-inflammatory and anti-tumor activity of FN and its effect on CAC, using biological fluid metabolomics to identify potential diagnostic markers for CAC. METHODS The MTT assay determined the survival rate of FN on murine RAW264.7 cells and the half maximal inhibitory concentration (IC50) of FN on murine CT-26 and human HCT116 cells IL-6, IL-1β, and TNF-α levels were detected by ELISA. Western blotting was used to analyze autophagic and apoptotic pathways. Utilizing an in vivo mouse model of colitis, the anti-inflammatory activity of FN was assessed by monitoring relevant indicators such as body weight, DAI score, and colon index. And an in vitro mouse colon cancer model was used to observe FN's anti-tumor activity by measuring tumor volume, size, and inhibition rate. Metabolomics analyzed differential serum metabolites and lipid metabolic pathways. Intestinal flora in mice was also analyzed. RESULTS FN can inhibit the activation of the NF-κB/MAPK signaling pathways in LPS-induced RAW264.7 cells, thereby exerting its anti-inflammatory effects. Moreover, FN significantly enhanced the colon length and DAI score in mice, notably suppressed the production of inflammatory cytokines, and inhibited the NF-κB/MAPK signaling pathway, leading to an improvement in DSS-induced colitis. FN significantly inhibited CT-26 and HCT116 cell growth, reduced tumor growth rate, improved pathological damage in CAC mice, and inhibited inflammatory factor production, enhancing intestinal mucosa protection. FN promoted apoptosis of colon cancer cells by increasing autophagy proteins (LC3, Beclin-1) and apoptosis proteins (CL-Caspase3, Bax), while reducing Bcl-2 expression. Metabolomics identified 34 differential metabolites, including glycerophospholipids and fatty acids, showing anti-tumor effects by regulating lipid metabolism. FN reduced IGF-1, ACLY, A-CoA, FAS, HSL, ATGL, and FFA levels, and increased GSK-3 levels (p < 0.01). FN also inhibited the expression of P-mTOR, Rictor, P-Akt, ACLY, PDE3B, P-PKA, and P-HSL (p < 0.01). CONCLUSIONS FN significantly inhibits colon cancer cell growth and exerts anti-CAC effects by activating autophagy and apoptosis pathways and regulating lipid metabolism. This study is the first to comprehensively integrate metabolomics, intestinal flora analysis, and molecular mechanisms to unveil FN's multifaceted role in CAC treatment, offering novel insights into its therapeutic application.
Collapse
Affiliation(s)
- Mingxiu Liu
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Canjiao Yang
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Xingju Peng
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Suman Zheng
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Hongping He
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Weiguang Wang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650031, China
| | - Yanping Li
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China.
| |
Collapse
|
4
|
Chen M, Fang H, Gao S, Zheng T, Kao S, Qin Y, Zhao X, Zhou X, Zhu B, Huang B. Establishment of GDF15 time-resolved fluorescence immunoassay and its clinical application in colorectal cancer. Anal Biochem 2025; 702:115848. [PMID: 40118237 DOI: 10.1016/j.ab.2025.115848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/14/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
OBJECTIVE This study aimed to develop a highly sensitive time-resolved fluoroimmunoassay for growth differentiation factor 15 (GDF15-TRFIA) and investigate its clinical applicability in colorectal cancer (CRC). METHODS Using the principle of double-antibody sandwich immunity, the GDF15-TRFIA was established by solid-phase capture antibody and labeled detection antibody with europium as a tracer, the levels of serum GDF15 were quantified in healthy controls (HCs) and patients, and the value of GDF15 in the diagnosis of CRC was analyzed. RESULTS The established method has a wide measurement range and good linearity. The HOOK effect was not observed when GDF15 was less than 2000 ng/mL. The intra-analytical coefficients of variation (CVs) were 3.27 %-4.54 %, and the inter-analytical CVs were 5.84 %-10.41 %, and recoveries were 88.15 %-112.36 %. The correlation between GDF15-TRFIA and ELISA was good (ρ = 0.9284). Serum GDF15 levels were significantly higher in CRC patients than in benign colorectal tumor (BCT) patients and HCs (P < 0.0001). ROC analysis showed that simultaneous detection of CEA, CA19-9, and GDF15 significantly improved the diagnostic efficiency of CEA and CA19-9. CONCLUSION A highly sensitive GDF15-TRFIA method for serum GDF15 was successfully established. It can be used for preliminary diagnosis of CRC, and expected to be a good auxiliary tool for the future clinical diagnosis of CRC.
Collapse
Affiliation(s)
- Meichun Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Hongming Fang
- Affiliated Xiao Shan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Shang Gao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Tianyu Zheng
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Shangbin Kao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xueqin Zhao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiumei Zhou
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.
| | - Bao Zhu
- Department of Nuclear Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, 214023, China.
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.
| |
Collapse
|
5
|
Pereira M, Brandão Ostermann RA, de Fáveri W, Damiani AP, Magenis ML, de Oliveira Monteiro I, Longaretti LM, Zaccaron RP, Lock Silveira PC, Bazo AP, Trevisan Frajacomo FT, Moraes de Andrade V. Vitamin C and D do not increase the chemopreventive effect of aspirin on colon carcinogenesis in a mouse model. Food Chem Toxicol 2025; 200:115400. [PMID: 40118136 DOI: 10.1016/j.fct.2025.115400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Aspirin has significant antineoplastic effects on the colon, whereas the outcomes of Vitamin C and D supplementation have been inconsistent. This study addressed the isolated and combined effects of vitamins C and D with aspirin on chemically induced colon carcinogenesis in mice, focusing on redox balance and DNA damage. Adult male and female mice were divided into seven groups: a control group; a group treated with 1,2-dimethylhydrazine (DMH, 40 mg/kg twice per week during weeks 4 and 5); and groups treated with DMH plus the following: aspirin (6 mg/kg in water); Vitamin C (500 mg/L in water); Vitamin D (1500 IU by oral gavage); a combination of Vitamin C and aspirin; and a combination of Vitamin D and aspirin. The treatments were administered continuously for 12 weeks. The treatments, isolated or combined, reduced aberrant crypt formations. Aspirin alone reduced the formation of aberrant crypt foci (ACF) by 65 %, accompanied by a systemic reduction in oxidative stress and DNA damage prevention. However, adding vitamins C and D to aspirin did not enhance these effects. Vitamin D alone suppressed ACF formation and DNA damage in the liver, whereas vitamin C had a limited effect on colon carcinogenesis, despite reducing oxidative stress in the liver and colon. In summary, we found no evidence that vitamins C and D supplementation enhanced the chemopreventive effects of aspirin on colon cancer.
Collapse
Affiliation(s)
- Maiara Pereira
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Rafael Alencastro Brandão Ostermann
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Wanessa de Fáveri
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Adriani Paganini Damiani
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Marina Lumertz Magenis
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Isadora de Oliveira Monteiro
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Luiza Martins Longaretti
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Rubya Pereira Zaccaron
- Laboratory of Experimental Pathophysiology, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Pathophysiology, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Ana Paula Bazo
- Center for Applied Studies in Health - Barriga Verde University Center - UNIBAVE, Orleans, SC, Brazil
| | | | - Vanessa Moraes de Andrade
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil.
| |
Collapse
|
6
|
Moskal K, Khurana N, Siegert L, Lee YS, Clevers H, Elinav E, Puschhof J. Modeling cancer-microbiome interactions in vitro: A guide to co-culture platforms. Int J Cancer 2025; 156:2053-2067. [PMID: 39716471 PMCID: PMC11970552 DOI: 10.1002/ijc.35298] [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: 07/15/2024] [Revised: 10/10/2024] [Accepted: 10/29/2024] [Indexed: 12/25/2024]
Abstract
The biology of cancer is characterized by an intricate interplay of cells originating not only from the tumor mass, but also its surrounding environment. Different microbial species have been suggested to be enriched in tumors and the impacts of these on tumor phenotypes is subject to intensive investigation. For these efforts, model systems that accurately reflect human-microbe interactions are rapidly gaining importance. Here we present a guide for selecting a suitable in vitro co-culture platform used to model different cancer-microbiome interactions. Our discussion spans a variety of in vitro models, including 2D cultures, tumor spheroids, organoids, and organ-on-a-chip platforms, where we delineate their respective advantages, limitations, and applicability in cancer microbiome research. Particular focus is placed on methodologies that facilitate the exposure of cancer cells to microbes, such as organoid microinjections and co-culture on microfluidic devices. We highlight studies offering critical insights into possible cancer-microbe interactions and underscore the importance of in vitro models in those discoveries. We anticipate the integration of more complex microbial communities and the inclusion of immune cells into co-culture systems to more accurately simulate the tumor microenvironment. The advent of ever more sophisticated co-culture models will aid in unraveling the mechanisms of cancer-microbiome interplay and contribute to exploiting their potential in novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Kamil Moskal
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
- DKFZ Hector Cancer Institute at the University Medical CenterMannheimGermany
| | - Nimisha Khurana
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Luisa Siegert
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
| | - Ye Seul Lee
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Hans Clevers
- Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC UtrechtHubrecht InstituteUtrechtThe Netherlands
- Present address:
Roche Pharmaceutical Research and Early DevelopmentBaselSwitzerland
| | - Eran Elinav
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Systems Immunology DepartmentWeizmann Institute of ScienceRehovotIsrael
| | - Jens Puschhof
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
- DKFZ Hector Cancer Institute at the University Medical CenterMannheimGermany
| |
Collapse
|
7
|
Song Q, Jin Z, Zhang H, Hong K, Zhu B, Yin H, Yu B. Fusobacterium nucleatum-derived 3-indolepropionic acid promotes colorectal cancer progression via aryl hydrocarbon receptor activation in macrophages. Chem Biol Interact 2025; 414:111495. [PMID: 40174685 DOI: 10.1016/j.cbi.2025.111495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/15/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
An increasing body of research indicates that Fusobacterium nucleatum (F. nucleatum) significantly influences the onset and progression of colorectal cancer (CRC). Our previous study has shown that F. nucleatum exerts pro-tumorigenic effects through aryl hydrocarbon receptor (AhR) activation. However, the role of its microbial metabolites in regulating immune responses remains unclear. Here, we report for the first time that F. nucleatum-derived 3-Indolepropionic acid (IPA) activates AhR in macrophages, driving M2 polarization and tumor-promoting immunosuppression. We discovered that culture supernatant of F. nucleatum (CSF) robustly activates AhR in macrophages. In co-culture systems, CSF upregulated the expression of the M2 marker CD206 and elevated mRNA levels of CD163, TGF-β, IL-10, and VEGF. In a subcutaneous allograft model, CSF induced an elevated number of CD206+ macrophages and decreased presence of CD8+ T cells within the tumor microenvironment, thereby promoting tumor growth. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) revealed IPA as a novel major AhR-activating metabolite in CSF. Strikingly, IPA recapitulated CSF's effects in promoting tumor cell migration and immunosuppression, both in vitro and in vivo. Critically, the AhR inhibitor CH223191 abolished both IPA-mediated M2 polarization and tumor growth. Our study revealed a novel mechanism by which F. nucleatum-derived IPA reprograms macrophages through AhR activation to fuel CRC progression, providing potential therapeutic targets for CRC treatment and prognosis improvement.
Collapse
Affiliation(s)
- Qi Song
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China; Key Laboratory of Hubei Province for Digestive System Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Zhiliang Jin
- Department of Oncology, The Second Clinical Medical College, Yangtze University, Jingzhou, 434000, Hubei Province, People's Republic of China
| | - Han Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China; Key Laboratory of Hubei Province for Digestive System Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Kunqiao Hong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China; Key Laboratory of Hubei Province for Digestive System Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Beibei Zhu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China; Key Laboratory of Hubei Province for Digestive System Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Haisen Yin
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.
| | - Baoping Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.
| |
Collapse
|
8
|
Gan QH, Li SQ, Gan XL, Jiang ZQ, Jian ZY. Parenteral nutrition with n-3 polyunsaturated fatty acids on nutrition inflammatory and immune status of gastrointestinal cancer patients: Meta-analysis. World J Gastrointest Surg 2025; 17:105743. [DOI: 10.4240/wjgs.v17.i5.105743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/18/2025] [Accepted: 04/08/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND The incidence of malignant tumors in the digestive system is increasing and is a threat to human health. However, the long duration from tumor detection to radical resection, stress responses due to surgical trauma, and insufficient nutritional intake increases the risk of malnutrition, immune function reduction, postoperative complications, and intestinal dysfunction among patients.
AIM To systematically investigate the association of parenteral nutrition enriched with n-3 polyunsaturated fatty acids (PUFAs) with the nutritional status of patients after gastrointestinal treatment.
METHODS Randomized controlled trials associated with PUFA-enriched parenteral nutrition administration in patients with digestive system malignancies were retrieved from online databases such as PubMed, EMBASE, ScienceDirect, Cochrane Library, China Knowledge Network, China VIP, Wanfang, and China Biomedical Literature Database, with the retrieval time from database inception to present. Two researchers independently extracted data. Each article’s bias risk was assessed by referring to the Cochrane Handbook version 5.3 criteria and RevMan5.4 was used for data analysis.
RESULTS This meta-analysis involved six randomized controlled trials involving a total of 505 cases. Random-effects model analysis indicated remarkably better improvements in various inflammatory factors in the study group (P < 0.05). Meta-analysis of nutritional indicators revealed that the study group had higher total protein, albumin, and prealbumin levels, as well as lower transferrin levels compared to the control group (P < 0.05). Meanwhile, meta-analysis of T-cell subsets revealed no remarkable inter-group difference in post-treatment CD8+ cells (P > 0.05). Moreover, the meta-analysis identified a notably lower incidence of adverse reactions in the study group (P < 0.05).
CONCLUSION Administration of PUFAs helps improve the nutritional status of patients with digestive malignancies in the perioperative period. It promotes immune function recovery, reduces the inflammatory response, and decreases the risk of adverse effects. These beneficial effects make it worth investigating and promoting their use in appropriate patient populations. However, further validation via high-quality studies with long intervention time and extended follow-up periods is required.
Collapse
Affiliation(s)
- Qin-Hu Gan
- Department of Gastrointestinal Surgery, Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Shu-Qun Li
- Department of Hepatobiliary and Pancreatic Surgery, Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Xin-Li Gan
- Department of Gastrointestinal Surgery, Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Qing Jiang
- Department of Gastrointestinal Surgery, Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Yuan Jian
- Department of Gastrointestinal Surgery, Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
9
|
Qian L, Yang Y, Zhao B, Xu P, Hu Z, Zhong L, Dai Q, Zhong Y, Yang C, Shu Q, Han RPS, Guan Y, Li Z, Chen L. Inhibition of colorectal carcinogenesis by sunitinib malate: disruption of the IL-6/STAT3/c-MYC/TWIST/MMP2 autocrine signaling axis. Discov Oncol 2025; 16:893. [PMID: 40410534 PMCID: PMC12102017 DOI: 10.1007/s12672-025-02498-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 04/25/2025] [Indexed: 05/25/2025] Open
Abstract
Sunitinib, a multi-targeted receptor tyrosine kinase inhibitor with specificity for VEGFR, KIT, FLT3, and PDGFR, has demonstrated clinical efficacy as a first- to third-line treatment for refractory renal carcinoma. Our previous research indicated that sunitinib malate suppresses intestinal polyp proliferation by downregulating IL-6 mRNA expression, suggesting a potential analogous mechanism in colorectal carcinoma inhibition. This study aimed to elucidate the pharmacological effects and molecular mechanisms of sunitinib malate on colorectal carcinoma using HCT116, RKO, HT29, and SW480 cell lines in vitro and HCT116-derived xenografts in nude mice in vivo. We employed a comprehensive array of experimental techniques, including CCK-8/MTT assays for cell viability, Transwell and/or wound healing assays for migration, and Western blot and immunohistochemistry for protein expression analysis. Our findings demonstrate that sunitinib malate significantly inhibits colorectal cancer cell proliferation and migration in vitro. Moreover, in the xenograft model, sunitinib malate markedly suppressed colorectal tumor growth in vivo. Notably, we observed significant downregulation of c-MYC, TWIST, and MMP2 expression both in vitro and in vivo following sunitinib malate treatment. These results collectively suggest that sunitinib malate exerts its anti-colorectal carcinoma effects, at least in part, by disrupting the autocrine IL-6/STAT3/c-MYC/TWIST/MMP2 signaling axis.
Collapse
Affiliation(s)
- Ling Qian
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, No. 1688, Meiling Road, Xinjian, Nanchang, 330004, Jiangxi, China
- The Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Nanchang, 330004, China
- Jiangxi Engineering Research Center for Translational Cancer Technology, Nanchang, 330004, China
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- Key Laboratory of Pharmacology of Chinese Medicine in Jiangxi, Nanchang, 330004, China
| | - Yi Yang
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, No. 1688, Meiling Road, Xinjian, Nanchang, 330004, Jiangxi, China
- The Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Nanchang, 330004, China
- Jiangxi Engineering Research Center for Translational Cancer Technology, Nanchang, 330004, China
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- Key Laboratory of Pharmacology of Chinese Medicine in Jiangxi, Nanchang, 330004, China
| | - Bin Zhao
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, No. 1688, Meiling Road, Xinjian, Nanchang, 330004, Jiangxi, China
- The Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Nanchang, 330004, China
- Jiangxi Engineering Research Center for Translational Cancer Technology, Nanchang, 330004, China
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- Key Laboratory of Pharmacology of Chinese Medicine in Jiangxi, Nanchang, 330004, China
| | - Pan Xu
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, No. 1688, Meiling Road, Xinjian, Nanchang, 330004, Jiangxi, China
- The Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Nanchang, 330004, China
- Jiangxi Engineering Research Center for Translational Cancer Technology, Nanchang, 330004, China
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- Key Laboratory of Pharmacology of Chinese Medicine in Jiangxi, Nanchang, 330004, China
| | - Ziyan Hu
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, No. 1688, Meiling Road, Xinjian, Nanchang, 330004, Jiangxi, China
- The Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Nanchang, 330004, China
- Jiangxi Engineering Research Center for Translational Cancer Technology, Nanchang, 330004, China
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- Key Laboratory of Pharmacology of Chinese Medicine in Jiangxi, Nanchang, 330004, China
| | - Liangwang Zhong
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, No. 1688, Meiling Road, Xinjian, Nanchang, 330004, Jiangxi, China
- The Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Nanchang, 330004, China
- Jiangxi Engineering Research Center for Translational Cancer Technology, Nanchang, 330004, China
- College of Life Sciences, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Qi Dai
- Hepatogastrosplenicobiliary Department, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Youbao Zhong
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Chao Yang
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, No. 1688, Meiling Road, Xinjian, Nanchang, 330004, Jiangxi, China
- The Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Nanchang, 330004, China
- Jiangxi Engineering Research Center for Translational Cancer Technology, Nanchang, 330004, China
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- Key Laboratory of Pharmacology of Chinese Medicine in Jiangxi, Nanchang, 330004, China
- College of Life Sciences, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Qinglong Shu
- College of Life Sciences, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Ray P S Han
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, No. 1688, Meiling Road, Xinjian, Nanchang, 330004, Jiangxi, China
- The Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Nanchang, 330004, China
- Jiangxi Engineering Research Center for Translational Cancer Technology, Nanchang, 330004, China
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- Key Laboratory of Pharmacology of Chinese Medicine in Jiangxi, Nanchang, 330004, China
| | - Yang Guan
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Zhiming Li
- The Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Nanchang, 330004, China.
- Oncology Department, Affiliated Hospital of Jiangxi University of Chinese Medicine, No. 445 Bayi Avenue, Nanchang, 330006, Jiangxi, China.
| | - Lai Chen
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, No. 1688, Meiling Road, Xinjian, Nanchang, 330004, Jiangxi, China.
- The Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Nanchang, 330004, China.
- Jiangxi Engineering Research Center for Translational Cancer Technology, Nanchang, 330004, China.
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
- Key Laboratory of Pharmacology of Chinese Medicine in Jiangxi, Nanchang, 330004, China.
- College of Life Sciences, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| |
Collapse
|
10
|
Dash P, Yadav V, Das B, Satapathy SR. Experimental toolkit to study the oncogenic role of WNT signaling in colorectal cancer. Biochim Biophys Acta Rev Cancer 2025:189354. [PMID: 40414319 DOI: 10.1016/j.bbcan.2025.189354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 05/19/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
Colorectal cancer (CRC) is linked to the WNT/β-catenin signaling as its primary driver. Aberrant activation of WNT/β-catenin signaling is closely correlated with increased incidence, malignancy, poorer prognosis, and even higher cancer-related death. Research over the years has postulated various experimental models that have facilitated an understanding of the complex mechanisms underlying WNT signaling in CRC. In the present review, we have comprehensively summarized the in vitro, in vivo, patient-derived, and computational models used to study the role of WNT signaling in CRC. We discuss the use of CRC cell lines and organoids in capturing the molecular intricacies of WNT signaling and implementing xenograft and genetically engineered mouse models to mimic the tumor microenvironment. Patient-derived models, including xenografts and organoids, provide valuable insights into personalized medicine approaches. Additionally, we elaborated on the role of computational models in simulating WNT signaling dynamics and predicting therapeutic outcomes. By evaluating the advantages and limitations of each model, this review highlights the critical contributions of these systems to our understanding of WNT signaling in CRC. We emphasize the need to integrate diverse model systems to enhance translational research and clinical applications, which is the primary goal of this review.
Collapse
Affiliation(s)
- Pujarini Dash
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Biswajit Das
- Department of Molecular Cell and Developmental Biology, University of California, Santa Cruz, USA
| | - Shakti Ranjan Satapathy
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
11
|
Chen X, Cao Y, Zhao Y, Ma Y, Shi X, Wang J, Jiang Z, Luo R, Deng Z, Zhou X, Yang J, Fu W. Neurodegeneration of local sympathetic inputs promotes colorectal cancer progression. Cancer Lett 2025; 625:217817. [PMID: 40409454 DOI: 10.1016/j.canlet.2025.217817] [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: 05/06/2025] [Revised: 05/12/2025] [Accepted: 05/21/2025] [Indexed: 05/25/2025]
Abstract
The nervous system can profoundly influence cancer prognosis, and this frontier of cancer neuroscience has increasingly garnered research attention. However, the involvement of neural signals in colorectal cancer remains incompletely understood. In this study, we exploit advanced three-dimensional imaging and conventional immunohistochemistry to observe a transitional loss of local sympathetic inputs from colorectal adenomas to adenocarcinomas in human patients. This negative correlation similarly occurs in the mouse models of colorectal cancer. Of importance, the pharmacologic ablation of sympathetic innervations significantly exaggerates the progression of colorectal tumors in the chemical-induced mouse model. We then demonstrate that the sympathetic neurotransmitter norepinephrine acts via α2-adrenergic receptors to elevate the cancer cell expression of chemokines to recruit CD8+ T cells, and the destruction of sympathetic signals leads to their reduction within the tumor microenvironment. Together, these results have elucidated a novel aspect of the neurodegeneration of local sympathetic inputs in promoting colorectal cancer progression.
Collapse
Affiliation(s)
- Xin Chen
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, 100191, China
| | - Ying Cao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Center for Life Sciences, IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Yifei Zhao
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, 100191, China
| | - Yao Ma
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, 100191, China
| | - Xiangchao Shi
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, 100191, China
| | - Junwei Wang
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, 100191, China
| | - Zhaoyu Jiang
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, 100191, China
| | - Renjie Luo
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, 100191, China
| | - Zhangyuzi Deng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Center for Life Sciences, IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Xin Zhou
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, 100191, China.
| | - Jing Yang
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, 100191, China; State Key Laboratory of Membrane Biology, School of Life Sciences, Center for Life Sciences, IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| | - Wei Fu
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
12
|
Wei XM, Lu SC, Li L, Gao YJ, Wang JY, Xi SY, Ye LYL, Shen WX, Wu MH, Duan DD, Cheng HB. Norcantharidin promotes M1 macrophage polarization and suppresses colorectal cancer growth. Acta Pharmacol Sin 2025:10.1038/s41401-025-01578-8. [PMID: 40394236 DOI: 10.1038/s41401-025-01578-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 04/27/2025] [Indexed: 05/22/2025]
Abstract
Colorectal cancer (CRC) is characterized by an immunosuppressive and inflammatory microenvironment, thus responds poorly to therapy. Previous studies show that norcantharidin (NCTD), a demethylated cantharidin (CTD) derived from Mylabris, exerts high efficacy in treating various cancers. In this study we investigated the antitumor effects of NCTD against CRC and the underlying mechanisms. Subcutaneous CRC models were established in balb/c mice using mouse colorectal cancer cell line CT26 and in balb/c nude mice using human colorectal cancer cell line HCT116. The mice were administered NCTD (2 or 4 mg·kg-1·d-1, i.p.) for 14 days. We showed that NCTD dose-dependently reduced the tumor growth in both the CRC models. Furthermore, NCTD markedly increased M1 macrophage infiltration in tumor tissue in both the CRC models. NCTD-induced macrophage M1 polarization was confirmed by flow cytometry and qPCR assays in both THP-1 cell-derived and RAW264.7 macrophage models in vitro. We demonstrated that NCTD (20, 40 μM) dose-dependently increased CSF2 secretion from CRC cells and macrophages, and suppressed the JAK2/STAT3 signaling pathway in CRC cells. Concurrently, NCTD (10-40 μM) dose-dependently inhibited CRC cell proliferation, invasion and migration in vitro. In conclusion, this study provides new evidence for the effects of NCTD against CRC and elucidates its antitumor mechanisms through remodeling the inflammatory microenvironment via CSF2-mediated macrophage M1 polarization and inhibiting JAK2/STAT3 phosphorylation in CRC cells.
Collapse
Affiliation(s)
- Xiao-Man Wei
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China
| | - Si-Cheng Lu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China
- School of Integrative Medicine of Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Liu Li
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China
| | - Ying-Jie Gao
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China
| | - Jun-Yi Wang
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Song-Yang Xi
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China
| | - Ling-Yu Linda Ye
- School of Integrative Medicine of Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wei-Xing Shen
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China.
| | - Mian-Hua Wu
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China.
| | - Dayue Darrel Duan
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China.
- Department of Pharmacology, University of Nevada Reno School of Medicine, Reno, NV, 89557, USA.
| | - Hai-Bo Cheng
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China.
| |
Collapse
|
13
|
Dal Secco C, Tonon S, Trevisan C, Martinis E, Valeri V, Codrich M, Tell G, Frossi B, Pucillo CEM. Mast cells-intestinal cancer cells crosstalk is mediated by TNF-alpha and sustained by the IL-33/ST2 axis. Cancer Immunol Immunother 2025; 74:205. [PMID: 40372523 DOI: 10.1007/s00262-025-04054-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 04/11/2025] [Indexed: 05/16/2025]
Abstract
It is common knowledge that mast cells (MCs) exert different roles in the gastrointestinal tract, from the maintenance of homeostasis to the onset and propagation of different gut diseases such as food allergies, infections, inflammation, and cancer. However, the mechanisms through which MCs dialog and influence the intestinal tissue are not completely known. To get insight into the bidirectional crosstalk between MCs and the intestinal microenvironment, both in homeostatic and pathological settings, colon organoids from intestinal epithelium of healthy mice and adenomas from AOM/DSS-treated mice have been exploited and co-cultured with MCs. The influence of MCs on organoid architecture and the effect of healthy and tumoral organoids on the phenotype and responsiveness of MCs have been addressed. We observed that MCs interact with intestinal organoids and contribute to the differentiation of healthy organoids by upregulating the expression of mucin-2, chromogranin A, cadherin-1, and claudin 4. On the contrary, in co-culture with tumoral organoids a decrease in cell proliferation, chromogranin A, and lysozyme expression was observed. Tumoral organoids have been shown to activate MCs via the IL-33/ST2 axis leading to increased release of TNF-α which in turn was responsible for the observed effects on tumoral organoids. Our results indicate that MCs are important mediators of intestinal tissue homeostasis and that a different environment can shape and direct MCs toward the dampening or propagation of the inflammatory response. Ultimately, our MC-organoid co-cultures represent a valid in vitro tool to investigate the role of MCs in the gut.
Collapse
Affiliation(s)
- Chiara Dal Secco
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Silvia Tonon
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Caterina Trevisan
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Eleonora Martinis
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Viviana Valeri
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Marta Codrich
- Molecular Biology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Gianluca Tell
- Molecular Biology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Barbara Frossi
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Carlo E M Pucillo
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy.
| |
Collapse
|
14
|
Lv Y, Mao W, Jin H, Qu J, He D. Associations of human exposure to 6PPD and 6PPDQ with colorectal cancer: A mixture analysis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 373:126114. [PMID: 40139299 DOI: 10.1016/j.envpol.2025.126114] [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: 01/17/2025] [Revised: 02/26/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine (6PPD) and its oxidation product, 6PPD-quinone (6PPDQ), are widely present in the environment. Toxicological studies have demonstrated that they can induce adverse health effects on the intestinal system. However, epidemiological studies examining the association between human 6PPD and 6PPDQ exposure and colorectal cancer (CRC) risk remain scarce. In this study, human urinary 6PPD and 6PPDQ concentrations were analyzed in 329 controls and 367 CRC cases from Quzhou, China. A combination of analyses, including unconditional logistic regression, Bayesian kernel machine regression (BKMR), and restricted cubic spline analysis, was employed to evaluate associations between urinary 6PPD and 6PPDQ levels and CRC risk, adjusting for demographic and lifestyle variables. The median concentration of 6PPDQ in CRC cases (0.94 vs 0.14 μg/g creatinine) was significantly higher than that in controls (Mann-Whitney U test, p = 0.001), while the median concentration of 6PPD showed no significant (p = 0.061) difference between the two groups (0.31 vs 0.38 μg/g creatinine). Higher urinary 6PPDQ concentrations were significantly associated with increased CRC risk, especially among participants with third (adjusted OR = 2.79, 95 % CI: 1.76-4.47; p for trend <0.001) and fourth (adjusted OR = 7.13, 95 % CI: 4.31-12.0; p for trend <0.001) quartiles of exposure. Additionally, the joint effects of 6PPD and 6PPDQ exposure, assessed using the BKMR model, indicated a positive association with CRC risk, suggesting a cumulative risk from co-exposure. This study provides the first epidemiological evidence linking human 6PPDQ exposure to CRC risk, highlighting its potential role in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Yangbo Lv
- Department of Colorectal Surgery, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, 324000, PR China
| | - Weili Mao
- Department of Pharmacy, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, 324000, PR China
| | - Hangbiao Jin
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang, 310032, PR China
| | - Jianli Qu
- College of Environmental Science and Engineering, Zhejiang University of Water Resources and Electric Power, Hangzhou, Zhejiang, 310018, PR China.
| | - Dongjuan He
- Department of Endocrinology, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, 324000, PR China.
| |
Collapse
|
15
|
Chen S, Jiang Z, Song W, Lu C, Lin Y, Xu S, Xie K, Wan L, Yuan X. Identification of the "Collagen-Macrophage" sub-category of patients with colorectal cancer as an extension of the CMS4 subtype with THBS2 as a therapeutic target. BMC Gastroenterol 2025; 25:342. [PMID: 40340827 PMCID: PMC12060322 DOI: 10.1186/s12876-025-03918-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 04/21/2025] [Indexed: 05/10/2025] Open
Abstract
We identified a subset of patients with colorectal cancer (CRC) enriched with "collagen-TAMs," designated the CM class, using large integrated colon cancer transcriptome and single-cell transcriptome datasets. This classification system could be used as an extension of the traditional CMS classification system for CRC to guide more accurate classification and treatment.We also screened CAF-derived THBS2 as a potential biomarker for CM and found that it plays an important role in CRC disease models in vitro and in vivo, promoting tumor development and metastasis as well as TAM recruitment. Targeting THBS2 combined with PD-1 therapy effectively improved the therapeutic effect of immunotherapy in vivo. The CM classification provides a new perspective for CRC treatment, and THBS2, which is highly expressed in CM cases, can be used as a new potential combined target for immunotherapy.
Collapse
Affiliation(s)
- Shuwen Chen
- Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, 211166, China
- Department of Clinical Medicine, First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China
| | - Zhaoyan Jiang
- Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, 211166, China
| | - Wanxuan Song
- Department of Clinical Medicine, First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China
| | - Chuqiao Lu
- Department of Clinical Medicine, First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China
| | - Yanbing Lin
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Shiyao Xu
- Department of Clinical Medicine, First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China
| | - Kunxin Xie
- Department of Biochemistry and Molecular Biology, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, China
| | - Li Wan
- Department of Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223302, China.
| | - Xiaoqin Yuan
- Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
16
|
Zhou H, Zhuang Y, Liang Y, Chen H, Qiu W, Xu H, Zhou H. Curcumin exerts anti-tumor activity in colorectal cancer via gut microbiota-mediated CD8 + T Cell tumor infiltration and ferroptosis. Food Funct 2025; 16:3671-3693. [PMID: 40244948 DOI: 10.1039/d4fo04045g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Colorectal cancer (CRC), as a high-incidence malignancy, continues to present significant challenges in prevention, screening, and treatment. Curcumin (Cur) exhibits notable anti-inflammatory and anticancer properties. Despite its poor solubility in water and low bioavailability, high concentrations of Cur are detected in the gastrointestinal tract after oral administration, suggesting that it may directly interact with the gut microbiota and exert regulatory effects. This study aims to explore the mechanisms by which Cur improves CRC by modulating gut microbiota. Firstly, we evaluated the effect of Cur on CRC cell viability in vitro using the MTT assay, and the results showed a significant inhibitory effect on CRC cell growth. The IC50 values for Cur in CT26 and RKO cells were 23.52 μM, 16.11 μM, and 13.62 μM at 24, 48, and 72 hours, respectively, and 26.3 μM, 16.52 μM, and 14.22 μM at 24, 48, and 72 hours, respectively. Cur induced apoptosis and caused G2 phase cell cycle arrest in tumor cells. Subsequently, we established a CRC mouse model. Oral administration of Cur at 15 mg kg-1 and 30 mg kg-1 inhibited CRC progression, as evidenced by reduced tumor volume, histological analysis, immunohistochemistry, and an increased number of CD8+ T cells infiltrating the tumors. Ferroptosis in tumor cells was also observed. Cur partially restored the gut microbiota of CRC mice, altering the abundance and diversity of the gut microbiota and affecting serum metabolite distribution, with significant increases in the abundance of SCFA-producing microbes such as Lactobacillus and Kineothrix. To verify causality, we designed a fecal microbiota transplantation (FMT) experiment. Compared with CRC mice, the fecal microbiota from Cur-treated mice significantly alleviated CRC symptoms, including slowed tumor growth, enhanced CD8+ T cell tumor infiltration, and induced ferroptosis in tumor cells. Additionally, when gut microbiota was depleted with antibiotics, Cur's antitumor effects disappeared, suggesting that Cur mitigates CRC in a gut microbiota-dependent manner. These findings provide new insights into the mechanisms underlying CRC and propose novel therapeutic interventions, emphasizing the interaction between gut microbiota and immune responses within the tumor immune microenvironment (TIME).
Collapse
Affiliation(s)
- Hongli Zhou
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, 210023, Nanjing, China
| | - Yupei Zhuang
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, 210023, Nanjing, China
| | - Yuwei Liang
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, 210023, Nanjing, China
| | - Haibin Chen
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
| | - Wenli Qiu
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, 210023, Nanjing, China
| | - Huiqin Xu
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
| | - Hongguang Zhou
- Nanjing University of Chinese Medicine, 210023, Nanjing, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, 210023, Nanjing, China
| |
Collapse
|
17
|
Bachet JB, de Gramont A, Raeisi M, Rakez M, Goldberg RM, Tebbutt NC, Van Cutsem E, Haller DG, Hecht JR, Mayer RJ, Lichtman SM, Benson AB, Sobrero AF, Tabernero J, Adams R, Zalcberg JR, Grothey A, Yoshino T, André T, Shi Q, Chibaudel B. Characteristics of Patients and Prognostic Factors Across Treatment Lines in Metastatic Colorectal Cancer: An Analysis From the Aide et Recherche en Cancérologie Digestive Database. J Clin Oncol 2025:JCO2401968. [PMID: 40324123 DOI: 10.1200/jco-24-01968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/22/2025] [Accepted: 03/14/2025] [Indexed: 05/07/2025] Open
Abstract
PURPOSE Several lines of treatment can be used sequentially in patients with metastatic colorectal cancer. We investigated the evolution of patient/tumor characteristics and their prognostic impact across treatment lines to develop an overall prognostic score (OPS). PATIENTS AND METHODS Individual patient data from 48 randomized trials were analyzed. The end point was overall survival (from random assignment to death). Missing data were imputed. The complete data set was then separated into construction (80%) and validation sets (20%). The Cox's model was used to define risk groups for survival using the OPS. The discrimination capability was assessed in each treatment-line via bootstrapping to obtain optimism-corrected calibration and discrimination C-indices. Internal validation was done in the validation set. RESULTS A total of 37,560 patients (26,974 in first-line [1L], 7,693 in second-line [2L], and 2,893 in third-line [3L]) were analyzed. Some clinical, biological, and molecular characteristics of patients/tumors included in therapeutic trials evolve over the lines. Seven independent prognostic variables were retained in the final multivariate model common to all lines: Eastern Cooperative Oncology Group performance status, hemoglobin, platelet count, WBC/absolute neutrophil count ratio, lactate dehydrogenase, alkaline phosphatase, and the number of metastatic sites. The OPS was used to define four patient subgroups with significantly different prognoses in 1L, 2L, and 3L, separately, with adequate C-indices: 0.65, 0.66, and 0.69 in the construction set and 0.65, 0.66, and 0.68 in the validation set, respectively. The OPS was not predictive, with 3L drugs (v placebo) or subsequent line (2L/1L or 3L/2L) extending survival in all prognostic groups. CONCLUSION The same prognostic model using practical variables can be used before all treatment lines. The OPS could better stratify patients in future clinical trials and help to therapeutic decision in routine practice.
Collapse
Affiliation(s)
- Jean-Baptiste Bachet
- Hepato-gastroenterology and Digestive Oncology Department, Pitié Salpêtrière Hospital, APHP, Sorbonne Université, Paris, France
| | - Aimery de Gramont
- Department of Medical Oncology, Franco-British Hospital, Fondation Cognacq-Jay, Cancérologie Paris Ouest, Levallois-Perret, France
- ARCAD Foundation, Paris, France
| | | | - Manel Rakez
- Statistical Unit, ARCAD Foundation, Paris, France
| | - Richard M Goldberg
- Department of Medicine, West Virginia University Cancer Institute, Morgantown, WV
| | - Niall C Tebbutt
- Department of Medical Oncology, Olivia Newton-John Cancer, Wellness and Research Centre, Austin Health, Heidelberg, VIC, Australia
| | - Eric Van Cutsem
- Digestive Oncology, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | - Daniel G Haller
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | | | | | - Stuart M Lichtman
- Wilmot Cancer Institute Geriatric Oncology Research Group, University of Rochester, Rochester, NY
| | - Al B Benson
- Division of Hematology/Oncology, Northwestern University's Feinberg School of Medicine, Chicago, IL
| | | | - Josep Tabernero
- Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology (VHIO), IOB-Quiron, Barcelona, Spain
| | - Richard Adams
- Cardiff University and Velindre Cancer Centre, Cardiff, United Kingdom
| | - John R Zalcberg
- Department of Medical Oncology, Monash University School of Public Health and Preventive Medicine, Alfred Health, Melbourne, VIC, Australia
| | | | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Thierry André
- ARCAD Foundation, Paris, France
- Department of Medical Oncology, Saint Antoine Hospital, APHP, Sorbonne Université, Paris, France
| | - Qian Shi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Benoist Chibaudel
- Department of Medical Oncology, Franco-British Hospital, Fondation Cognacq-Jay, Cancérologie Paris Ouest, Levallois-Perret, France
| |
Collapse
|
18
|
Maruyama Y, Hosonuma M, Toyoda H, Funayama E, Sasaki A, Baba Y, Tajima K, Nakashima R, Sasaki A, Shida M, Tsurui T, Tsuji M, Tsunoda T, Shimane T, Kiuchi Y, Yoshimura K, Kuramasu A. Combination therapy with cetirizine and anti-PD-1 antibody suppresses colitis-induced colon tumor formation in mice. Eur J Pharmacol 2025; 999:177704. [PMID: 40320111 DOI: 10.1016/j.ejphar.2025.177704] [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: 12/08/2024] [Revised: 04/24/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have transformed cancer therapy, yet their efficacy remains limited in inflammation-associated tumors, necessitating combinatorial approaches. Although combinations with cytotoxic or targeted agents are well established, the therapeutic potential of non-oncologic drugs, such as antihistamines, is less explored. In this study, we investigate whether cetirizine, a non-sedating histamine H1 receptor antagonist, enhances the antitumor effects of anti-programmed cell death protein 1 (PD-1) antibody in a murine model of colitis-associated colorectal cancer. Combination therapy, not monotherapy, significantly reduced tumor volume in vivo. Flow cytometry of splenocytes revealed increased PD-1 expression on T cells only in the combination group, suggesting systemic immune activation. Immunohistochemical analysis showed elevated CD3+ T-cell infiltration into tumors following combination treatment. Meanwhile, gene expression analysis of tumor tissues revealed downregulated Vegfa, Mmp9, Il10, and Cd80, along with upregulated Hspg2 and Fn1, suggesting a shift in the tumor microenvironment. In vitro, cetirizine suppressed Mmp9 expression in CT26 cells, Il10 in macrophages, and VEGFA in human umbilical vein endothelial cells, indicating cell-type-specific effects that partially mirror the in vivo findings. Immunohistochemistry further demonstrated a reduced frequency of FoxP3+ regulatory T cells among CD3+ T cells within the tumor stroma in the combination group. Collectively, these findings indicate that cetirizine enhances ICI efficacy by reshaping the tumor microenvironment through immunomodulatory mechanisms. Our results support the repurposing of antihistamines as a novel strategy to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Yuki Maruyama
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan; Department of Otorhinolaryngology-Head and Neck Surgery, Showa Medical University School of Medicine, Tokyo, Japan
| | - Masahiro Hosonuma
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan; Division of Medical Oncology, Department of Medicine, Showa Medical University School of Medicine, Tokyo, Japan
| | - Hitoshi Toyoda
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan; Department of Orthopaedic Surgery, School of Medicine, Showa Medical University, Tokyo, Japan
| | - Eiji Funayama
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan; Division of Pharmacology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa Medical University, Tokyo, Japan
| | - Akiko Sasaki
- Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan
| | - Yuta Baba
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan
| | - Kohei Tajima
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan
| | - Rie Nakashima
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan
| | - Aya Sasaki
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan
| | - Midori Shida
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan
| | - Toshiaki Tsurui
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan; Division of Medical Oncology, Department of Medicine, Showa Medical University School of Medicine, Tokyo, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa Medical University, Tokyo, Japan
| | - Takuya Tsunoda
- Division of Medical Oncology, Department of Medicine, Showa Medical University School of Medicine, Tokyo, Japan
| | - Toshikazu Shimane
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Head and Neck Oncology Center, Showa Medical University, Tokyo, Japan
| | - Yuji Kiuchi
- Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan
| | - Kiyoshi Yoshimura
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Division of Medical Oncology, Department of Medicine, Showa Medical University School of Medicine, Tokyo, Japan
| | - Atsuo Kuramasu
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan.
| |
Collapse
|
19
|
Buckenmeyer MJ, Brooks EA, Taylor MS, Orenuga IK, Yang L, Holewinski RJ, Meyer TJ, Galloux M, Garmendia-Cedillos M, Pohida TJ, Andresson T, Croix B, Wolf MT. A 3D Self-Assembly Platform Integrating Decellularized Matrix Recapitulates In Vivo Tumor Phenotypes and Heterogeneity. Cancer Res 2025; 85:1577-1595. [PMID: 39888317 PMCID: PMC12048290 DOI: 10.1158/0008-5472.can-24-1954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/17/2024] [Accepted: 01/24/2025] [Indexed: 02/01/2025]
Abstract
Three-dimensional (3D) in vitro cell culture models are invaluable tools for investigating the tumor microenvironment. However, analyzing the impact of critical stromal elements, such as extracellular matrix (ECM), remains a challenge. In this study, we developed a hydrogel-free self-assembly platform to establish ECM-rich 3D "MatriSpheres" to deconvolute cancer cell-ECM interactions. Mouse and human colorectal cancer MatriSpheres actively incorporated microgram quantities of decellularized small intestine submucosa ECM, which proteomically mimicked colorectal cancer tumor ECM compared with traditional formulations like Matrigel. Solubilized ECM, at subgelation concentrations, was organized by colorectal cancer cells into intercellular stroma-like regions within 5 days, displaying morphologic similarity to colorectal cancer clinical pathology. MatriSpheres featured ECM-dependent transcriptional and cytokine profiles associated with malignancy, lipid metabolism, and immunoregulation. Model benchmarking with single-cell RNA sequencing demonstrated that MatriSpheres enhanced correlation with in vivo tumor cells over traditional ECM-poor spheroids. This facile approach enables tumor-specific tissue morphogenesis, promoting cell-ECM communication to improve fidelity for disease modeling applications. Significance: MatriSpheres provide a hydrogel-free 3D platform for decoupling the influence of heterogeneous extracellular matrix components on tumor biology and can broadly facilitate high-throughput drug discovery and screening applications. See related commentary by Ernst and De Wever, p. 1568.
Collapse
Affiliation(s)
- Michael J. Buckenmeyer
- Cancer Biomaterials Engineering Laboratory, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Elizabeth A. Brooks
- Cancer Biomaterials Engineering Laboratory, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Madison S. Taylor
- Cancer Biomaterials Engineering Laboratory, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Ireolu K. Orenuga
- Cancer Biomaterials Engineering Laboratory, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Liping Yang
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Ronald J. Holewinski
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21701, USA
| | - Thomas J. Meyer
- CCR Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mélissa Galloux
- Independent Bioinformatician, Marseille, Provence-Alpes-Côte d’Azur, France
| | - Marcial Garmendia-Cedillos
- Instrumentation Development and Engineering Application Solutions, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Thomas J. Pohida
- Instrumentation Development and Engineering Application Solutions, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Thorkell Andresson
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21701, USA
| | - Brad Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Matthew T. Wolf
- Cancer Biomaterials Engineering Laboratory, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| |
Collapse
|
20
|
Sun X, Tong J, Fang X, Lu M, Rao C, Li Y. Comprehensive Multi-Omics Analysis of Copper Metabolism Related Molecular Subtypes and Prognostic Risk Stratification in Colon Adenocarcinoma. J Cell Mol Med 2025; 29:e70591. [PMID: 40391581 PMCID: PMC12089994 DOI: 10.1111/jcmm.70591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/21/2025] [Accepted: 04/26/2025] [Indexed: 05/22/2025] Open
Abstract
Colon adenocarcinoma (COAD) is the most common subtype of colorectal cancer, originating from glandular cells in the colon. Despite diagnostic and therapeutic advances, its prognosis remains poor. Copper, an essential micronutrient, is involved in tumorigenesis and other biological processes. In this study, we identified copper metabolism-related genes (CMRG) associated with COAD prognosis from TCGA and GEO databases and constructed a CMRG-based risk model. We assessed its clinical relevance through analyses of immune infiltration, immunotherapy response, and drug sensitivity. Single-cell sequencing revealed the spatial and cellular distribution of CMRG in COAD tissues, providing insight into their roles in the tumour microenvironment. COX19 was selected for further validation, and in vitro experiments (western blot, PCR, siRNA, colony formation, and Transwell assays) confirmed its role in promoting COAD cell invasion and proliferation. These findings highlight the involvement of copper metabolism in COAD progression and suggest potential targets for therapy.
Collapse
Affiliation(s)
- Xi Sun
- Department of Anorectal SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouChina
| | - Jingfei Tong
- Department of Anorectal SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouChina
| | - Xiaojie Fang
- Department of Anorectal SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouChina
| | - Miaojiong Lu
- Department of Anorectal SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouChina
| | - Chunhui Rao
- Department of Anorectal SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yanyan Li
- Department of Anorectal SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouChina
| |
Collapse
|
21
|
Zhong H, Jiang J, Hussain M, Zhang H, Chen L, Guan R. The Encapsulation Strategies for Targeted Delivery of Probiotics in Preventing and Treating Colorectal Cancer: A Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500304. [PMID: 40192333 PMCID: PMC12079478 DOI: 10.1002/advs.202500304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/01/2025] [Indexed: 05/16/2025]
Abstract
Colorectal cancer (CRC) ranks as the third most prevalent cancer worldwide. It is associated with imbalanced gut microbiota. Probiotics can help restore this balance, potentially reducing the risk of CRC. However, the hostile environment and constant changes in the gastrointestinal tract pose significant challenges to the efficient delivery of probiotics to the colon. Traditional delivery methods are often insufficient due to their low viability and lack of targeting. To address these challenges, researchers are increasingly focusing on innovative encapsulation technologies. One such approach is single-cell encapsulation, which involves applying nanocoatings to individual probiotic cells. This technique can improve their resistance to the harsh gastrointestinal environment, enhance mucosal adhesion, and facilitate targeted release, thereby increasing the effectiveness of probiotic delivery. This article reviews the latest developments in probiotic encapsulation methods for targeted CRC treatment, emphasizing the potential benefits of emerging single-cell encapsulation techniques. It also analyzes and compares the advantages and disadvantages of current encapsulation technologies. Furthermore, it elucidates the underlying mechanisms through which probiotics can prevent and treat CRC, evaluates the efficacy and safety of probiotics in CRC treatment and adjuvant therapy, and discusses future directions and potential challenges in the targeted delivery of probiotics for CRC treatment and prevention.
Collapse
Affiliation(s)
- Hao Zhong
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
| | - Jin Jiang
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
| | - Muhammad Hussain
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
- Moganshan Institute ZJUTKangqianDeqing313200China
| | - Haoxuan Zhang
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
| | - Ling Chen
- Sanya Branch of Hainan Academy of Inspection and TestingSan Ya572011China
| | - Rongfa Guan
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
- Moganshan Institute ZJUTKangqianDeqing313200China
| |
Collapse
|
22
|
Rahimi A, Baghernejadan Z, Hazrati A, Malekpour K, Samimi LN, Najafi A, Falak R, Khorramdelazad H. Combination therapy with immune checkpoint inhibitors in colorectal cancer: Challenges, resistance mechanisms, and the role of microbiota. Biomed Pharmacother 2025; 186:118014. [PMID: 40157004 DOI: 10.1016/j.biopha.2025.118014] [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: 01/08/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Colorectal cancer (CRC) is still one of the leading causes of cancer deaths worldwide. Even though there has been progress in cancer immunotherapy, the results of applying immune checkpoint inhibitors (ICIs) have been unsatisfactory, especially in microsatellite stable (MSS) CRC. Single-agent ICIs that target programmed cell death-1 (PD-1)/ PD-L1, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), T cell Ig- and mucin-domain-containing molecule-3 (TIM-3), and lymphocyte activation gene (LAG)-3 have emerged as having specific benefits. However, many primary and secondary resistance mechanisms are available in the tumor microenvironment (TME) that prevent it from happening. Combination strategies, such as the use of anti-PD-1 and anti-CTLA-4, can be effective in overcoming these resistance pathways, but toxicities remain a significant concern. Moreover, ICIs have been integrated with various treatment modalities, including chemotherapy, radiotherapy, antibiotics, virotherapy, polyadenosine diphosphate-ribose polymerase (PARP) inhibitors, and heat shock protein 90 (HSP90) inhibitors. The outcomes observed in both preclinical and clinical settings have been encouraging. Interestingly, manipulating gut microbiota via fecal microbiota transplantation (FMT) has been identified as a new strategy to increase the efficacy of immunotherapy in CRC patients. Therefore, integrating ICIs with other treatment approaches holds promise in enhancing the prognosis of CRC patients. This review focuses on the unmet need for new biomarkers to select patients for combination therapies and the ongoing work to overcome resistance and immune checkpoint blockade.
Collapse
Affiliation(s)
- Ali Rahimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Baghernejadan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alireza Najafi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
23
|
Long M, Chen W, Li J, Kong W, Su M, Qiu L, Cheng X, Bi L. Redox-responsive chondroitin sulfate-based micelle system for enhanced chemotherapy and inflammation suppression to synergistically antitumor therapy. Int J Biol Macromol 2025; 311:143686. [PMID: 40311976 DOI: 10.1016/j.ijbiomac.2025.143686] [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: 12/30/2024] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/03/2025]
Abstract
According to the close association between cancer and inflammation demonstrated in clinical data, the strategy of synergistic anti-tumor and anti-inflammatory therapy shows promising potential. However, challenges remain in the synthesis and development of co-delivery systems for synergistic therapy. Herein, an amphiphilic chondroitin sulfate-rosmarinic acid polymeric prodrug was synthesized, and then combined with DSPE-PEG to encapsulate doxorubicin, forming a redox-responsive nanomicelle (PRSC/DOX) delivery system. PRSC/DOX exhibited a particle size of 188.6 nm and remained stable in PBS for at least 7 days. PRSC/DOX was internalized into tumor cells via CD44 receptor-mediated endocytosis, and degraded by intracellular glutathione to release the drugs. The released doxorubicin killed tumor cells through chemotherapy, and rosmarinic acid inhibited tumor cell growth by suppressing inflammation levels in the tumor microenvironment. In vivo experiments showed a statistically significant decrease in the inflammation levels in mice, along with a considerable reduction in tumor volume. Consequently, the PRSC/DOX significantly enhanced antitumor efficacy through a synergistic therapy of chemotherapy and inflammation suppression.
Collapse
Affiliation(s)
- Miaomiao Long
- Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing 210042, China; Department of Pharmacy, Wuxi Higher Health Vocational Technology School, Wuxi 214028, China
| | - Weijun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Jie Li
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Weibo Kong
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Min Su
- Hunan Clinical Medical Research Center of Accurate Diagnosis and Treatment for Esophageal Carcinoma, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China; Hunan RunKun Pharmaceutical Co., Ltd., Yuehang 414003, China.
| | - Lipeng Qiu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Xian Cheng
- Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing 210042, China.
| | - Liangwu Bi
- Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing 210042, China.
| |
Collapse
|
24
|
Herranz-Montoya I, Angulo-Aguado M, Perna C, Zagorac S, García-Jimeno L, Park S, Djouder N. p53 protein degradation redefines the initiation mechanisms and drives transitional mutations in colorectal cancer. Nat Commun 2025; 16:3934. [PMID: 40287431 PMCID: PMC12033273 DOI: 10.1038/s41467-025-59282-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Incidence of colorectal cancer (CRC) is increasing likely due to different mechanisms driving initiation and progression. The initial model proposed by Fearon and Vogelstein posits it as a multi-hit neoplasia, originating from adenomatous-polyps induced by WNT activation, ultimately progressing to aggressiveness through p53 loss. Integrating human data with mouse genetics, we redefine this paradigm, highlighting pivotal roles of MYC, oncogenic URI and p53 degradation to initiate CRC. Early APC loss activates MYC to transcriptionally upregulate URI, which modulates MDM2 activity, triggering p53 proteasomal degradation, essential for tumour initiation and mutation burden accrual in CRC mice. Remarkably, reinstating p53 levels via genetic URI depletion or p53 super-expression in CRC mice with WNT pathway activation prevents tumour initiation and extends lifespan. Our data reveal a "two-hit" genetic model central to APC loss-driven CRC initiation, wherein MYC/URI axis intricately controls p53 degradation, offering mechanistic insights into transitional mutation acquisition essential for CRC progression.
Collapse
Affiliation(s)
- Irene Herranz-Montoya
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), 28029, Madrid, Spain
| | - Mariana Angulo-Aguado
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), 28029, Madrid, Spain
| | - Cristian Perna
- Department of Pathology, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
- Universidad de Alcalá, 28801, Madrid, Spain
| | - Sladjana Zagorac
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), 28029, Madrid, Spain
| | - Luis García-Jimeno
- Computational Cancer Genomics Group, Structural Biology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), 28029, Madrid, Spain
| | - Solip Park
- Computational Cancer Genomics Group, Structural Biology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), 28029, Madrid, Spain
| | - Nabil Djouder
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), 28029, Madrid, Spain.
| |
Collapse
|
25
|
Li KJ, Zhang ZY, Wang K, Sulayman S, Zeng XY, Liu J, Chen Y, Zhao ZL. Prognostic scoring system using inflammation- and nutrition-related biomarkers to predict prognosis in stage I-III colorectal cancer patients. World J Gastroenterol 2025; 31:104588. [PMID: 40248373 PMCID: PMC12001188 DOI: 10.3748/wjg.v31.i14.104588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/22/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a common malignancy that has become a global burden. The prognostic prediction of CRC patients on the basis of inflammatory biomarkers and nutritional biomarkers has shown some potential but has not been fully explored. AIM To develop and validate a prognostic model for CRC based on inflammation and nutrition-related biomarkers and to evaluate its predictive value for patient outcomes. METHODS Patients were randomized at a 3:2 ratio into a training cohort (n = 282) or a validation cohort (n = 188). To identify the optimal prognostic factors for constructing the risk score (RS), LASSO Cox regression analysis was conducted. The association between the RS and overall survival (OS) was evaluated using receiver operating characteristic (ROC) curves and Kaplan-Meier (K-M) survival analysis. Independent risk factors were screened by multivariate Cox regression analysis. Nomograms were constructed and validated on the basis of these factors. RESULTS In the training cohort, univariate analysis of all the inflammatory and nutritional biomarkers demonstrated some predictive value. A LASSO-Cox analysis included four biomarkers and constructed an RS. Through ROC analysis, the area under the prognostic curve was 0.795. K-M survival curve analyses revealed that the five-year OS was significantly greater in the Low-RS group than in the High-RS group (P < 0.001). Multivariate analysis demonstrated that the degree of differentiation (P = 0.001), degree of nerve invasion (P = 0.022), and RS (P < 0.001) were independent risk factors. We constructed a nomogram to predict the OS of CRC patients and validated it in a separate cohort. The calibration curve showed high accuracy. Additionally, decision curve analysis for 1-year, 3-year, and 5-year survival probabilities indicated significant clinical utility in predicting survival outcomes. CONCLUSION This study developed a nomogram based on the RS to predict the OS of CRC patients. This nomogram can guide treatment decisions and enable the formulation of personalized follow-up strategies on the basis of predicted recurrence risk, aiming to improve long-term prognosis.
Collapse
Affiliation(s)
- Ke-Jin Li
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Zi-Yi Zhang
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Kuan Wang
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Subinur Sulayman
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Xiang-Yue Zeng
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Juan Liu
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Yi Chen
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Ze-Liang Zhao
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
26
|
Tang J, Chen L, Shen X, Xia T, Li Z, Chai X, Huang Y, Yang S, Peng X, Lai J, Li R, Xie L. Exploring the Role of Cellular Interactions in the Colorectal Cancer Microenvironment. J Immunol Res 2025; 2025:4109934. [PMID: 40255905 PMCID: PMC12008489 DOI: 10.1155/jimr/4109934] [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: 08/23/2024] [Accepted: 02/22/2025] [Indexed: 04/22/2025] Open
Abstract
Colorectal cancer (CRC) stands as one of the tumors with globally high incidence and mortality rates. In recent years, researchers have extensively explored the role of the tumor immune microenvironment (TME) in CRC, highlighting the crucial influence of immune cell populations in driving tumor progression and shaping therapeutic outcomes. The TME encompasses an array of cellular and noncellular constituents, spanning tumor cells, immune cells, myeloid cells, and tumor-associated fibroblasts, among others. However, the cellular composition within the TME is highly dynamic, evolving throughout different stages of tumor progression. These shifts in cell subpopulation proportions lead to a gradual transition in the immune response, shifting from an early antitumor growth to a late-stage environment that supports tumor survival. Therefore, it is crucial to further investigate and understand the complex interactions among the various cell populations within the TME. In this review, we explore the key cellular components of varying origins, subpopulations with shared origins, and noncellular elements within the CRC TME, examining their interconnections and critical considerations for developing personalized and precise immunotherapy strategies.
Collapse
Affiliation(s)
- Jiadai Tang
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Liuhan Chen
- Department of Head and Neck Surgery Section II, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Xin Shen
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Tingrong Xia
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Zhengting Li
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Xiaoying Chai
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Yao Huang
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Shaoqiong Yang
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Xinjun Peng
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Junbo Lai
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Rui Li
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Lin Xie
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| |
Collapse
|
27
|
Lushnikova A, Wickbom A, Bohr J, Kruse R, Wirén A, Hultgren Hörnquist E. Increased Colonic Levels of CD8+ Cytotoxic T lymphocyte-Associated Mediators in Patients With Microscopic Colitis. Inflamm Bowel Dis 2025:izaf064. [PMID: 40209110 DOI: 10.1093/ibd/izaf064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Indexed: 04/12/2025]
Abstract
BACKGROUND For unidentified reasons, possibly due to increased immune surveillance, patients with collagenous colitis (CC) and lymphocytic colitis (LC), both forms of microscopic colitis (MC), have lower risk of colorectal cancer than controls and ulcerative colitis (UC) patients. Levels of secreted and cell-bound mediators in MC patients with active disease and in histological remission (HR) compared to UC patients and controls were investigated. METHODS Median fluorescence intensity of 54 analytes in colonic biopsies from patients with active CC (n = 21), LC (n = 11), and UC (n = 19); CC-HR (n = 6), LC-HR (n = 9), UC in remission (n = 19), non-diarrhea controls (n = 48), and diarrhea controls (n = 25) was measured using Luminex. RESULTS Granzyme B and CCL5 levels were higher in active CC than in UC, whereas CCL4 and CD163 levels were similar in CC and UC, and both groups had higher levels of matrix metalloproteinase (MMP)-1, MMP-3, and tumor necrosis factor receptor II than both control groups. APRIL, BAFF, BCMA, CCL20, CXCL8, chitinase 3-like 1, pentraxin-3, Fas, and IL-33 were higher in UC than MC. Increases in 4-1BB and perforin in MC compared to controls were lower than in UC. Levels of gp130 and IL-6Rα were decreased in MC but increased in UC compared to controls. CONCLUSIONS Microscopic colitis patients exhibit increased levels of several analytes, including some associated with CD8+ T lymphocytes, suggesting a different pathogenesis of MC compared to UC. Higher levels of MMP-1 and MMP-3 in CC than LC indicate separate disease entities.
Collapse
Affiliation(s)
| | - Anna Wickbom
- Division of Gastroenterology, Department of Medicine, Örebro University Hospital, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Johan Bohr
- Division of Gastroenterology, Department of Medicine, Örebro University Hospital, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Robert Kruse
- School of Medical Sciences, Örebro University, Örebro, Sweden
- Department of Clinical Research Laboratory, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Anders Wirén
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | | |
Collapse
|
28
|
Papadimitriou N, Kazmi N, Tsilidis KK, Richmond RC, Lynch BM, Bendinelli B, Ricceri F, Sánchez MJ, Trobajo-Sanmartín C, Jakszyn P, Simeon V, Severi G, Perduca V, Truong T, Ferrari P, Keski-Rahkonen P, Weiderpass E, Eichelmann F, Schulze MB, Katzke V, Fortner RT, Heath AK, Aune D, Harewood R, Dahm CC, Llorente A, Gunter MJ, Murphy N, Lewis SJ. Identifying Metabolomic Mediators of the Physical Activity and Colorectal Cancer Relationship. Cancer Epidemiol Biomarkers Prev 2025; 34:578-587. [PMID: 39883068 PMCID: PMC11966109 DOI: 10.1158/1055-9965.epi-24-1390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/27/2024] [Accepted: 01/28/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Current evidence suggests higher physical activity (PA) levels are associated with a reduced risk of colorectal cancer. However, the mediating role of the circulating metabolome in this relationship remains unclear. METHODS Targeted metabolomics data from 6,055 participants in the European Prospective Investigation into Cancer and Nutrition cohort were used to identify metabolites associated with PA and derive a metabolomic signature of PA levels. PA levels were estimated using the validated Cambridge PA index based on baseline questionnaires. Mediation analyses were conducted in a nested case-control study (1,585 cases, 1,585 controls) to examine whether individual metabolites and the metabolomic signature mediated the PA-colorectal cancer association. RESULTS PA was inversely associated with colorectal cancer risk (OR per category change: 0.90, 95% confidence interval, 0.83-0.97; P value = 0.009). PA levels were associated with 24 circulating metabolites after FDR correction, with the strongest associations observed for phosphatidylcholine acyl-alkyl (PC ae) C34:3 (FDR-adjusted P value = 1.18 × 10-10) and lysophosphatidylcholine acyl C18:2 (FDR-adjusted P value = 1.35 × 10-6). PC ae C34:3 partially mediated the PA-colorectal cancer association (natural indirect effect: 0.991, 95% confidence interval, 0.982-0.999; P value = 0.04), explaining 7.4% of the association. No mediation effects were observed for the remaining metabolites or the overall PA metabolite signature. CONCLUSIONS PC ae C34:3 mediates part of the PA-colorectal cancer inverse association, but further studies with improved PA measures and extended metabolomic panels are needed. IMPACT These findings provide insights into PA-related biological mechanisms influencing colorectal cancer risk and suggest potential targets for cancer prevention interventions.
Collapse
Affiliation(s)
- Nikos Papadimitriou
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Nabila Kazmi
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, The Medical School, University of Bristol, Bristol, United Kingdom
| | - Konstantinos K. Tsilidis
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
- Cancer Epidemiology and Prevention Research Unit, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Rebecca C. Richmond
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, The Medical School, University of Bristol, Bristol, United Kingdom
| | - Brigid M. Lynch
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
- Physical Activity Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Benedetta Bendinelli
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Cinical Network (ISPRO), Florence, Italy
| | - Fulvio Ricceri
- Department of Clinical and Biological Sciences, Centre for Biostatistics, Epidemiology, and Public Health (C-BEPH), University of Turin, Turin, Italy
| | - Maria-Jose Sánchez
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Camino Trobajo-Sanmartín
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Instituto de Salud Pública y Laboral de Navarra, Pamplona, Spain
- Navarre Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Paula Jakszyn
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
- Blanquerna School of Health Sciences, Ramon Llull University, Barcelona, Spain
| | - Vittorio Simeon
- Unit of Medical Statistics, Department of Mental and Physical Health and Preventive Medicine, University of Campania, Naples, Italy
| | - Gianluca Severi
- Department of Statistics, Computer Science, Applications “G. Parenti”, University of Florence, Florence, Italy
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, CESP, Villejuif, France
| | - Vittorio Perduca
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, CESP, Villejuif, France
- Université Paris Cité, CNRS, MAP5, Paris, France
| | - Therese Truong
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, CESP, Villejuif, France
| | - Pietro Ferrari
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Pekka Keski-Rahkonen
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | | | - Fabian Eichelmann
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Matthias B. Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Verena Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Renée Turzanski Fortner
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | - Alicia K. Heath
- Cancer Epidemiology and Prevention Research Unit, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Dagfinn Aune
- Cancer Epidemiology and Prevention Research Unit, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
- Department of Nutrition, Oslo New University College, Oslo, Norway
| | - Rhea Harewood
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Christina C. Dahm
- Department of Public Health, Aarhus University, Aarhus Centrum, Denmark
| | - Adrian Llorente
- Subdirection of Public Health of Guipuzcoa, San Sebastián, Spain
| | - Marc J. Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
- Cancer Epidemiology and Prevention Research Unit, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Neil Murphy
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Sarah J. Lewis
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, The Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
29
|
Ding Y, Yu Y. Therapeutic potential of flavonoids in gastrointestinal cancer: Focus on signaling pathways and improvement strategies (Review). Mol Med Rep 2025; 31:109. [PMID: 40017144 PMCID: PMC11884236 DOI: 10.3892/mmr.2025.13474] [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: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
Flavonoids are a group of polyphenolic compounds distributed in vegetables, fruits and other plants, which have considerable antioxidant, anti‑tumor and anti‑inflammatory activities. Several types of gastrointestinal (GI) cancer are the most common malignant tumors in the world. A large number of studies have shown that flavonoids have inhibitory effects on cancer, and they are recognized as a class of potential anti‑tumor drugs. Therefore, the present review investigated the molecular mechanisms of flavonoids in the treatment of different types of GI cancer and summarized the drug delivery systems commonly used to improve their bioavailability. First, the classification of flavonoids and the therapeutic effects of various flavonoids on human diseases were briefly introduced. Then, to clarify the mechanism of action of flavonoids on different types of GI cancer in the human body, the metabolic process of flavonoids in the human body and the associated signaling pathways causing five common types of GI cancer were discussed, as well as the corresponding therapeutic targets of flavonoids. Finally, in clinical settings, flavonoids have poor water solubility, low permeability and inferior stability, which lead to low absorption efficiency in vivo. Therefore, the three most widely used drug delivery systems were summarized. Suggestions for improving the bioavailability of flavonoids and the focus of the next stage of research were also put forward.
Collapse
Affiliation(s)
- Ye Ding
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yong Yu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
30
|
Zhang Y, Abousamra S, Hasan M, Torre-Healy L, Krichevsky S, Shrestha S, Bremer E, Oldridge DA, Rech AJ, Furth EE, Bocklage TJ, Levens JS, Hands I, Durbin EB, Samaras D, Kurc T, Saltz JH, Gupta R. Pathomics Image Analysis of Tumor Infiltrating Lymphocytes (TILs) in Colon Cancer. RESEARCH SQUARE 2025:rs.3.rs-6173056. [PMID: 40235501 PMCID: PMC11998795 DOI: 10.21203/rs.3.rs-6173056/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
We developed a deep learning Pathomics image analysis workflow to generate spatial Tumor-TIL maps to visualize and quantify the abundance and spatial distribution of tumor infiltrating lymphocytes (TILs) in colon cancer. Colon cancer and lymphocyte detection in hematoxylin and eosin (H&E) stained whole slide images (WSIs) has revealed complex immuno-oncologic interactions that form TIL-rich and TIL-poor tumor habitats, which are unique in each patient sample. We compute Tumor%, total lymphocyte%, and TILs% as the proportion of the colon cancer microenvironment occupied by intratumoral lymphocytes for each WSI. Kaplan-Meier survival analyses and multivariate Cox regression were utilized to evaluate the prognostic significance of TILs% as a Pathomics biomarker. High TILs% was associated with improved overall survival (OS) and progression-free interval (PFI) in localized and metastatic colon cancer and other clinicopathologic variables, supporting the routine use of Pathomics Tumor-TIL mapping in biomedical research, clinical trials, laboratory medicine, and precision oncology.
Collapse
|
31
|
Liu W, Lau HCH, Ding X, Yin X, Wu WKK, Wong SH, Sung JJY, Zhang T, Yu J. Transmission of antimicrobial resistance genes from the environment to human gut is more pronounced in colorectal cancer patients than in healthy subjects. IMETA 2025; 4:e70008. [PMID: 40236771 PMCID: PMC11995172 DOI: 10.1002/imt2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 04/17/2025]
Abstract
Antimicrobial resistance is a major global health concern. However, the source of gut resistome remains unsolved. We aimed to analyze the contribution of environmental antimicrobial resistance genes (ARGs) to colorectal cancer (CRC) patients. Here, we collected metagenomic data from 1,605 human stool samples (CRC = 748; healthy = 857) and 1,035 city-matched environmental samples, in which 110 CRC, 112 healthy, and 56 environmental samples were newly collected. Compared to healthy subjects, CRC patients had significantly higher ARG burden (p < 0.01) with increased levels of multidrug-resistant ARGs. Gut ARGs in CRC also had a closer similarity to environmental ARGs (p < 0.001). By comparing environmental and gut ARGs, 28 environmental ARGs were identified as CRC-specific ARGs, including SUL2 and MEXE, which were not identified in healthy subjects. Meanwhile, more mobile ARGs (mARGs) from the environment were observed in CRC patients compared to healthy subjects (p < 0.05). The hosts of mARGs were mainly pathogenic bacteria (e.g., Escherichia coli (E. coli) and Clostridium symbiosum (C. symbiosum)). Compared to healthy subjects, CRC patients showed elevated horizontal gene transfer efficiency from the environment to gut. Consistently, the abundance of pathobionts carrying specific mARGs (e.g., E. coli-SUL2 and C. symbiosum-SUL2) were significantly increased in CRC patients compared to healthy subjects (p < 0.05). We thus reveal a route of ARG dissemination from the environment into the gut of CRC patients.
Collapse
Affiliation(s)
- Weixin Liu
- Institute of Digestive Disease and The Department of Medicine and TherapeuticsState Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong KongHong Kong SARChina
| | - Harry C. H. Lau
- Institute of Digestive Disease and The Department of Medicine and TherapeuticsState Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong KongHong Kong SARChina
| | - Xiao Ding
- Institute of Digestive Disease and The Department of Medicine and TherapeuticsState Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong KongHong Kong SARChina
| | - Xiaole Yin
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil EngineeringThe University of Hong KongHong Kong SARChina
| | - William Ka Kei Wu
- Institute of Digestive Disease and The Department of Medicine and TherapeuticsState Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong KongHong Kong SARChina
| | - Sunny Hei Wong
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Joseph J. Y. Sung
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Tong Zhang
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil EngineeringThe University of Hong KongHong Kong SARChina
| | - Jun Yu
- Institute of Digestive Disease and The Department of Medicine and TherapeuticsState Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong KongHong Kong SARChina
| |
Collapse
|
32
|
Hauan M, Rylander C, Skeie G. Sweet beverages and the risk of colorectal cancer: the Norwegian Women and Cancer Study. BMC Cancer 2025; 25:592. [PMID: 40169938 PMCID: PMC11963648 DOI: 10.1186/s12885-025-13835-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common type of cancer worldwide, with Norwegian women having the highest incidence rate of colon cancer in 2022. The consumption of sweet beverages is a suggested modifiable risk factor for CRC; however, current evidence is limited and inconclusive. OBJECTIVE To assess the associations between the intake of sugar-sweetened beverages (SSBs), artificially sweetened beverages (ASBs), and juice and the risk of overall and subsite-specific CRC among Norwegian women. METHODS In this prospective cohort study, we included 73,921 participants aged 41-61 years at baseline. Information on sweet beverage consumption was collected using self-reported food frequency questionnaires at two time points between 1998 and 2014. We used Cox proportional hazards models to estimate hazard ratios (HRs) with corresponding 95% confidence intervals (CIs) for the associations between sweet beverage consumption and the risk of overall CRC, proximal colon cancer, distal colon cancer, and rectal cancer. RESULTS During a mean follow-up time of 16.5 years from baseline, 1,187 women were diagnosed with CRC. Compared to no consumption, juice consumption was inversely associated with overall CRC risk (HR≥ 7 glasses/wk = 0.81, 95% CI: 0.67-0.98; p-trend = 0.025), colon cancer (HR≥ 7 glasses/wk = 0.73, 95% CI: 0.58-0.94; p-trend = 0.015) and proximal colon cancer (HR≥ 7 glasses/wk = 0.71, 95% CI: 0.52-0.99; p-trend = 0.065) after adjusting for age, education, and diabetes status at baseline. No associations were observed between juice consumption and distal colon cancer or rectal cancer risk, or between the intake of SSBs or ASBs and CRC. CONCLUSION We observed no substantial association between the intake of SSBs or ASBs and the risk of CRC or cancer in colorectal subsites in our cohort of Norwegian women. Conversely, our results indicate that juice consumption is associated with a reduced risk of CRC, particularly in the colon. These results warrant further investigation in larger cohorts with power to detect possible differences in cancer risk across colorectal subsites, especially as patterns of sweet beverage consumption are changing.
Collapse
Affiliation(s)
- Marie Hauan
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway.
| | - Charlotta Rylander
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Guri Skeie
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
- IARC Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
33
|
He KJ, Gong G. Deciphering the integrated immunogenomic landscape of colorectal cancer: insights from Mendelian randomization and immune-stratified molecular subtyping. BMC Gastroenterol 2025; 25:213. [PMID: 40165100 PMCID: PMC11959972 DOI: 10.1186/s12876-025-03776-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
PURPOSE This study aimed to decipher the intricate interplay between the immune landscape and CRC pathogenesis, elucidating how distinct immunophenotypes causally influence disease susceptibility and stratify patient outcomes. METHODS We obtained the immunocyte phenotypes and CRC data from their respective genome-wide association studies. The primary analysis used the inverse variance weighting (IVW) method. We also simultaneously employed MR-Egger, weighted mode, simple mode, and weighted median approaches to strengthen the findings. Consensus clustering stratified 619 TCGA CRC patients by immunome expression. Functional assays examined the tumor suppressor GPD1L. RESULTS The IVW MR analysis identified 17 immunocyte phenotypes positively potentially associated with increased CRC risk (P < 0.05, OR > 1), and 18 phenotypes negatively potentially associated with decreased CRC risk (P < 0.05, OR < 1). These associations were not confounded by heterogeneity or horizontal pleiotropy (P > 0.05). Reverse MR analysis further revealed 4 additional immunocyte phenotypes positively potentially associated with CRC (P < 0.05, OR > 1). Clustering resolved prognostic C1/C2 subtypes dependent on coordinated immunophenotypic programs. GPD1L knockdown promoted CRC cell proliferation. CONCLUSIONS Genetic interrogation delineated causal immunome-CRC relationships at single-cell resolution. Immune-stratified CRC subtyping stratified patient outcomes. GPD1L exhibited tumor-suppressive functions. Our findings establish an integrated immunogenomic framework elucidating CRC pathogenesis with implications for precision immunotherapies.
Collapse
Affiliation(s)
- Ke-Jie He
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou city, Zhejiang Province, China.
| | - Guoyu Gong
- School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
34
|
Naveed M, Mughal MS, Aziz T, Makhdoom SI, Jamil H, Ali Khan A, Al-Hoshani N, Al-Joufi FA, Tahir Kassim RM, Alwethaynani MS. Exploration of mRNA-modifying METTL3 oncogene as momentous prognostic biomarker responsible for colorectal cancer development. Open Med (Wars) 2025; 20:20251167. [PMID: 40177651 PMCID: PMC11964186 DOI: 10.1515/med-2025-1167] [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: 04/19/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Background Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide, emphasizing the need for improved prognostic biomarkers. Recent studies have identified the mRNA-modifying METTL3 oncogene as a potential biomarker in CRC progression. Objective This study aimed to investigate the expression patterns of METTL3 in CRC, assess its association with clinical outcomes, identify interacting proteins and biological pathways, and explore its correlation with immune cell infiltration. Methods Comprehensive analyses were conducted using public datasets, including transcriptome profiles from The Cancer Genome Atlas and the GSE103512 dataset. Protein-protein interaction (PPI) networks, pathway enrichment, and immune infiltration analyses were performed to elucidate METTL3's role in CRC progression. Results METTL3 expression was significantly higher in CRC tissues compared to normal tissues (p < 0.001). Mutations in METTL3 were detected in approximately 6% of CRC cases, with fusion events involving the SRPK2 gene. PPI analysis identified ten interacting proteins, including METTL4, EIF3H, RBM15B1, CBLL1, WTAP, NCBP1, RBM15, ZC3H13, METTL14, and KIAA1429. METTL3 expression showed a positive correlation with METTL4, METTL14, NCBP1, and WTAP expression (R > 0.5, p < 0.001). Higher METTL3 expression was associated with immunosuppressive phenotypes, such as increased infiltration of tumor-associated macrophages, regulatory T cells, and cancer-associated fibroblasts (p < 0.001). Pathway enrichment analysis revealed METTL3's involvement in crucial pathways, including the cell cycle and renal cell carcinoma (p < 0.01). Gene ontology analysis highlighted its role in mRNA and RNA-related processes. Conclusion The study supports the potential of METTL3 as a prognostic biomarker in CRC and highlights its involvement in immune modulation and cancer progression. These findings lay the groundwork for future studies aimed at developing targeted therapies and improving patient outcomes.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Muhammad Saad Mughal
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Tariq Aziz
- Laboratory of Animal Health Food Hygiene and Quality, University of Ioannina, Arta, 47132, Greece
| | - Syeda Izma Makhdoom
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Hamza Jamil
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Ayaz Ali Khan
- Department of Biotechnology, University of Malakand, Chakdara, 18800, Pakistan
| | - Nawal Al-Hoshani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Fakhria A. Al-Joufi
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Aljouf, Saudi Arabia
| | | | - Maher S. Alwethaynani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Alquwayiyah, Riyadh, Saudi Arabia
| |
Collapse
|
35
|
Qin Y, Wang Q, Lin Q, Liu F, Pan X, Wei C, Chen J, Huang T, Fang M, Yang W, Pan L. Multi-omics analysis reveals associations between gut microbiota and host transcriptome in colon cancer patients. mSystems 2025; 10:e0080524. [PMID: 40013792 PMCID: PMC11915798 DOI: 10.1128/msystems.00805-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 01/31/2025] [Indexed: 02/28/2025] Open
Abstract
Colon cancer (CC) is one of the most common cancers globally, which is associated with the gut microbiota intimately. In current research, exploring the complex interaction between microbiomes and CC is a hotspot. However, the information on microbiomes in most previous studies is based on fecal, which does not fully display the microbial environment of CC. Herein, we collected mucosal and tissue samples from both the tumor and normal regions of 19 CC patients and clarified the composition of mucosal microbiota by 16S rRNA and metagenomic sequencing. Additionally, RNA-Seq was also conducted to identify the different expression genes between tumor and normal tissue samples. We revealed significantly different microbial community structures and expression profiles to CC. Depending on correlation analysis, we demonstrated that 1,472 genes were significantly correlated with CC tumor microbiota. Our study reveals a significant enrichment of Campylobacter jejuni in the mucosa of CC, which correlates with bile secretion. Additionally, we observe a negative correlation between C. jejuni and immune cells CD4+ Tem and mast cells. Finally, we discovered that metabolic bacterial endosymbiont of Bathymodiolus sp., Bacillus wiedmannii, and Mycobacterium tuberculosis had a significant survival value for CC, which was ignored by previous research. Overall, our study expands the understanding of the complex interplay between microbiota and CC and provides new targets for the treatment of CC. IMPORTANCE This study contributes to our understanding of the interaction between microbiota and colon cancer (CC). By examining mucosal and tissue samples rather than solely relying on fecal samples, we have uncovered previously unknown aspects of CC-associated microbiota. Our findings reveal distinct microbial community structures and gene expression profiles correlated with CC progression. Notably, the enrichment of Campylobacter jejuni in CC mucosa, linked to bile secretion, underscores potential mechanisms in CC pathogenesis. Additionally, observed correlations between microbial taxa and immune cell populations offer new avenues for immunotherapy research in CC. Importantly, this study introduces CC-associated microbiota with survival implications for CC, expanding therapeutic targets beyond conventional strategies. By elucidating these correlations, our study not only contributes to uncovering the potential role of gut microbiota in colon cancer but also establishes a foundation for mechanistic studies of gut microbiota in colon cancer, emphasizing the broader impact of microbiota research on cancer biology.
Collapse
Affiliation(s)
- Yuling Qin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiang Wang
- Guangxi Clinical Research Center for Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiumei Lin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fengfei Liu
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaolan Pan
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Caibiao Wei
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Junxian Chen
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Taijun Huang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Min Fang
- Guangxi Clinical Research Center for Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Weilong Yang
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Institute of Advanced Biotechnology and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Linghui Pan
- Guangxi Clinical Research Center for Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
36
|
Gu C, Sha G, Zeng B, Cao H, Cao Y, Tang D. Therapeutic potential of fecal microbiota transplantation in colorectal cancer based on gut microbiota regulation: from pathogenesis to efficacy. Therap Adv Gastroenterol 2025; 18:17562848251327167. [PMID: 40104324 PMCID: PMC11915259 DOI: 10.1177/17562848251327167] [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: 11/25/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related deaths worldwide, with its progression intricately linked to gut microbiota dysbiosis. Disruptions in microbial homeostasis contribute to tumor initiation, immune suppression, and inflammation, establishing the microbiota as a key therapeutic target. Fecal microbiota transplantation (FMT) has emerged as a transformative approach to restore microbial balance, enhance immune responses, and reshape the tumor microenvironment. This review explores the mechanisms underlying FMT's therapeutic potential, evaluates its advantages over other microbiota-based interventions, and addresses challenges such as donor selection, safety concerns, and treatment standardization. Looking forward, the integration of FMT into personalized CRC therapies requires robust clinical trials and the identification of predictive biomarkers to optimize its efficacy and safety.
Collapse
Affiliation(s)
- Chen Gu
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Gengyu Sha
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Binbin Zeng
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Herong Cao
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yibo Cao
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dong Tang
- Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou 225000, China
- The Yangzhou Clinical College of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, 221000, China
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People’s Hospital, Yangzhou University, Yangzhou, 225000, China
- Northern Jiangsu People’s Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| |
Collapse
|
37
|
Ao X, Zhou X, Liu J, Wu Q, Yang Y, Liu Y, Hao W, Li L, Wang K, Li Z. Insect medicines for colorectal cancer: A review of mechanisms, preclinical evidence, and future prospects. Medicine (Baltimore) 2025; 104:e41873. [PMID: 40101066 PMCID: PMC11922444 DOI: 10.1097/md.0000000000041873] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
Colorectal cancer is recognized as the third most prevalent malignant tumor globally. The recommended treatment modalities, including surgery, radiotherapy, and chemotherapy, are frequently associated with severe side effects and high recurrence rates. Cancer experts are actively engaged in a global pursuit of safer and more efficacious treatment strategies for colorectal cancer (CRC). Insect medicine, a unique subset of traditional Chinese medicine, is characterized by their broad spectrum of therapeutic effects, which include antibacterial, anticoagulant, antithrombotic, and sedative actions. Insects are enriched with proteins, peptides, and amino acids. These compounds exhibit pharmacological activities, including anti-tumor effects, inhibition of cancer cell proliferation, induction of apoptosis in cancer cells, anti-inflammatory properties, and immunomodulation. Recent studies have revealed that certain traditional Chinese insect medicines, such as Bombyx Batryticatus, Tubiechong, and Aspongopus chinensis Dalls, demonstrate outstanding therapeutic efficacy in the treatment of CRC. The anti-CRC actions of these insect medicines are potentially mediated through mechanisms involving the Hedgehog and Wnt/β-catenin signaling pathways, as well as immunomodulatory effects. Consequently, these insect medicines are proposed as a potential strategy for CRC treatment.
Collapse
Affiliation(s)
- Xinyi Ao
- Department of Spleen and Stomach Diseases, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, the Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xin Zhou
- Department of Spleen and Stomach Diseases, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, the Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jianqin Liu
- Department of Spleen and Stomach Diseases, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, the Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Qian Wu
- Department of Spleen and Stomach Diseases, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, the Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Yanlin Yang
- Department of Spleen and Stomach Diseases, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, the Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Yali Liu
- Department of Spleen and Stomach Diseases, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, the Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Weian Hao
- Department of Spleen and Stomach Diseases, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, the Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Li Li
- Department of Spleen and Stomach Diseases, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, the Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Kaixuan Wang
- Department of Spleen and Stomach Diseases, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhi Li
- Department of Spleen and Stomach Diseases, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, the Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
38
|
Silinskaite U, Valciukiene J, Jakubauskas M, Poskus T. The Immune Environment in Colorectal Adenoma: A Systematic Review. Biomedicines 2025; 13:699. [PMID: 40149674 PMCID: PMC11940254 DOI: 10.3390/biomedicines13030699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Research on colorectal adenoma is significantly less comprehensive compared to studies on colorectal carcinoma. Although colorectal adenoma is a precursor of the majority of sporadic colorectal cancers, not all adenomas develop into carcinomas. The complex interaction of immune responses in the premalignant tumor microenvironment might be a factor for that. Methods: In this systematic review, we aim to provide a thorough analysis of the current research examining the immune infiltration patterns in sporadic colorectal adenoma tissues in the context of immune cell-based, cytokine-based, and other immunological factor-related changes along the conventional adenoma-carcinoma sequence. The articles included in the review extend up to December 2024 in PubMed and Web of Science databases. Results: Most included studies have shown significant differences in immune cell counts, densities, and cytokine expression levels associated with premalignant colorectal lesions (and/or colorectal cancer). No consensus on the immune-related tendencies concerning CD4+T cells and CD8+T cells was reached. Decreasing expression of mDCs and plasma and naïve B cells were detected along the ACS. The increased density of tissue eosinophils in the adenoma tissue dramatically diminishes after the transition to carcinoma. As the adenoma progresses, the increasing expression of IL-1α, IL-4, IL-6, IL-8, IL-10, IL-17A, IL-21, IL-23, IL-33, and TGF-β and decreasing levels of IL-12A, IL-18, IFN-γ, and TNFα cytokines in the invasive carcinoma stage is being detected. The over-expression of COX-2, PD-1/PD-L1, CTLA-4, and ICOS/ICOSLG in the colorectal adenomatous and cancerous tissues was also observed. Conclusions: Further studies are needed for a better understanding of the whole picture of colorectal adenoma-associated immunity and its impact on precancerous lesion's potential to progress.
Collapse
|
39
|
Denk D, Ramakrishnan M, Conche C, Pallangyo C, Pesic M, Ceteci F, Kennel KB, Kirisözü AC, Engel E, Mohs K, Ritter B, Pardo AM, Özkurt E, Hildebrand F, Waisman A, Arkan MC, Greten FR. IL-17RA signaling provides dual tumor-suppressor function during late-stage colorectal carcinogenesis. Immunity 2025; 58:701-715.e8. [PMID: 40023157 DOI: 10.1016/j.immuni.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 09/13/2024] [Accepted: 02/05/2025] [Indexed: 03/04/2025]
Abstract
Expression of interleukin (IL)-17 family cytokines is associated with tumor-promoting inflammation. We found that low expression of IL17RA associated with worse prognosis in late-stage colorectal cancer (CRC) patients. Deletion of Il17ra in intestinal epithelial cells (IECs) in a murine model of CRC enhanced epithelial-to-mesenchymal transition (EMT) via increased expression of the epidermal growth factor receptor and subsequent activation of the kinase Src. Yet, these mice were protected from metastatic disease; Il17ra deletion impaired intestinal barrier function and enhanced systemic fungal invasion and associated immunity. However, in macrophages, IL-17RA was required for spleen tyrosine kinase (Syk) activation upon fungal-induced dectin-1 engagement, and Il17ra ablation impaired IL-18 release and protective CD8+ T cell-mediated anti-tumor immunity. Combining recombinant IL-17 and heat-killed Candida albicans rendered colorectal tumors sensitive to α-PD-1 treatment in a model of microsatellite stable (MSS) CRC. Thus, IL-17RA engages two distinct tumor-suppressive mechanisms in CRC, linking EMT and fungal-induced anti-tumor immunity during tumor progression.
Collapse
Affiliation(s)
- Dominic Denk
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany; Goethe University Frankfurt, University Hospital, Medical Clinic 1, 60590 Frankfurt/Main, Germany
| | - Mallika Ramakrishnan
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany
| | - Claire Conche
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany
| | - Charles Pallangyo
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany
| | - Marina Pesic
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany
| | - Fatih Ceteci
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany
| | - Kilian B Kennel
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany
| | - Asude C Kirisözü
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany
| | - Esther Engel
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany
| | - Kathleen Mohs
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany
| | - Birgit Ritter
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany
| | - Angeles Macias Pardo
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany
| | - Ezgi Özkurt
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK; Decoding Biodiversity, Earlham Institute, Norwich NR4 7UZ, Norfolk, UK
| | - Falk Hildebrand
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK; Decoding Biodiversity, Earlham Institute, Norwich NR4 7UZ, Norfolk, UK
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Melek C Arkan
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Frankfurt Cancer Institute, Goethe University Frankfurt, 60596 Frankfurt/Main, Germany
| | - Florian R Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Frankfurt Cancer Institute, Goethe University Frankfurt, 60596 Frankfurt/Main, Germany.
| |
Collapse
|
40
|
Guo Y, Shang S, Liang L, Liu E. ZNF385A was identified as a novel colorectal cancer-related functional gene by analysis of the interaction and immune characteristics of oxidative stress and the inflammatory response. Discov Oncol 2025; 16:290. [PMID: 40064736 PMCID: PMC11893970 DOI: 10.1007/s12672-025-02024-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Recently, oxidative stress and inflammatory responses have been shown to directly impact tumor growth and the tumor microenvironment (TME). However, more research is necessary to fully understand the relationship between oxidative stress and inflammatory responses and colorectal cancer (CRC). METHODS The FindCluster algorithm was used to extract CRC Single-cell RNA sequencing (scRNA-seq) data and identify tumor cell groupings. From the MSigDB database, genes associated with oxidative stress and the inflammatory response were taken. We identified molecular subtypes and built a predictive risk model with the LASSO-Cox method using the ConsensusClusterPlus software suite. We incorporated the prognostic risk model and other clinicopathological parameters into a column-line chart. Finally, we used Quantitative Polymerase Chain Reaction (qPCR) and immunohistochemistry to check the expression of the unreported hub model genes. Cell proliferation was assessed using EDU and colony formation assays. Reactive Oxygen Species (ROS) tests were used to quantitatively determine the ROS content in CRC cells. The ability of CRC cells to invade and migrate was examined using transwell experiments. The regulatory functions of hub model genes were discovered in vivo using a xenograft model tumor assay. RESULTS Oxidative stress and inflammatory response factors in monocytic/macrophages of CRC were significantly upregulated, and their oxidative stress and inflammatory response functions were significantly higher than those of other cell subgroups, as indicated by the enrichment score. These factors showed significant synergistic overexpression and enrichment in this cell population. We constructed a prognostic risk model consisting of seven signatures. The good and stable prognostic evaluation efficacy of the model was confirmed, and risk scores were determined to be independent prognostic factors for CRC. We explored the relationship between the risk score model and malignant progression of tumor cells, tumor immune microenvironment, genomic variation, chemotherapy resistance, and immune response. Further qPCR and immunohistochemistry analysis showed that the expression of ZNF385A was high in CRC tissues. The functional experiment results indicated that interfering with the expression of ZNF385A could suppress the proliferation, ROS, migration and invasion of SW620 cells in vitro and the growth of xenograft tumors in vivo. CONCLUSION In this study, we investigated the critical expression patterns of oxidative stress- and inflammatory response-related genes in CRC, which may contribute to the prognosis and immunotherapy of CRC. Additionally, we discovered ZNF385A to be a novel oncogene in CRC. These findings imply that this model may be applied to assess prognostic risk and identify potential therapeutic targets for CRC patients.
Collapse
Affiliation(s)
- Yaqi Guo
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Shipeng Shang
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Leilei Liang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Enrui Liu
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China.
| |
Collapse
|
41
|
Xiong H, He Z, Ding J, Liu J, Xue Y, Ji M, Hu N, Wu K, Deng X, Liu Z, Luo T, Deng X. Long‑term exposure of polyethylene nanoplastics promotes colorectal tumorigenesis. ENVIRONMENTAL CHEMISTRY LETTERS 2025. [DOI: 10.1007/s10311-025-01829-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 02/17/2025] [Indexed: 04/02/2025]
|
42
|
Sui Y, Hoshi N, Okamoto N, Inoue Y, Funatsu T, Ku Y, Ooi M, Watanabe D, Miyazaki H, Agawa M, Nakamura H, Ohgaki R, Kanai Y, Yang H, Kodama Y. The role of LAT1 in AOM/DSS-induced colorectal tumorigenesis. Biochem Biophys Res Commun 2025; 751:151446. [PMID: 39922055 DOI: 10.1016/j.bbrc.2025.151446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Amino acid transporters are essential for supplying nutrients to cells and are implicated in tumor progression. L-type amino acid transporter 1 (LAT1) is reported to be overexpressed in various cancers, affecting tumor development. However, the exact mechanisms by which LAT1 affects colorectal cancer (CRC) arising from a chronic inflammatory background are not yet fully understood. This study aimed to explore the role of LAT1 in CRC. Mice with intestinal epithelium-specific deletions of LAT1 (LAT1fl/fl; vil-cre) were treated with azoxymethane (AOM)/dextran sulfate sodium (DSS) in a colitis-associated cancer (CAC) model. Our results demonstrated that LAT1 was detected in normal colon crypts and highly expressed in AOM/DSS-induced tumor tissue. During the chronic colitis phase, weight loss was more prominent in LAT1fl/fl; vil-cre mice, compared with that in LAT1fl/fl mice. IL-1β and IL-6 expressions significantly increased in LAT1-deleted tumors; however, no overall difference in colon tumor number or size was observed between LAT1fl/fl and LAT1fl/fl; vil-cre mice. Accordingly, cell proliferation and apoptotic cell number were similar when comparing LAT1-deleted tumors with those with sufficient LAT1. Our findings indicated that LAT1 might not phenotypically affect overall colonic tumor development in this model; however, it affected the chronic colitis phase and inflammatory status within the tumors. These findings suggest that severe inflammation in tumors might have compensated for tumor growth in defects of amino acid supplementation through LAT1 deficiency, and provide insights into the potential of LAT1-targeted therapies for clinical CRC treatment.
Collapse
Affiliation(s)
- Yunlong Sui
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China; Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Namiko Hoshi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan; Division of Integrated Analyses of Bioresource and Health Care, Kobe University Graduate School of Medicine, Hyogo, 650-0047, Japan.
| | - Norihiro Okamoto
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Yuta Inoue
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Takumi Funatsu
- Division of Integrated Analyses of Bioresource and Health Care, Kobe University Graduate School of Medicine, Hyogo, 650-0047, Japan
| | - Yuna Ku
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Makoto Ooi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Daisuke Watanabe
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Haruka Miyazaki
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Misaki Agawa
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Hirotaka Nakamura
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Ryuichi Ohgaki
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, 565-0871, Japan
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, 565-0871, Japan; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka, 565-0871, Japan
| | - Hui Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| |
Collapse
|
43
|
Yao L, Gu C, Ge R, Zhang X, Meng X, Wang L, Peng D, Li G. Acetylated Dendrobium huoshanense polysaccharide: a novel inducer of apoptosis in colon cancer cells via Fas-FasL pathway activation and metabolic reprogramming. Front Oncol 2025; 15:1529868. [PMID: 40104499 PMCID: PMC11913854 DOI: 10.3389/fonc.2025.1529868] [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: 02/12/2025] [Indexed: 03/20/2025] Open
Abstract
Introduction Not all polysaccharides function as antitumor drugs, nor do they universally possess the same advantages regarding safety and biocompatibility. Those polysaccharides that are effective antitumor agents typically demonstrate superior safety profiles and biocompatibility compared to synthetic anticancer drugs, which can exhibit high toxicity and harmful side effects. Dendrobium huoshanense polysaccharide (DHP) has been recognized for its potential bioactive properties, particularly in anti-tumor treatment. This study investigates the effects of DHP on the proliferation and apoptosis of HCT116 colon cancer cells. Methods DHP was extracted according to previously published experimental methods. The inhibitory effects of DHP were evaluated using IEC6, Caco-2, and HCT116 cell lines, with changes in cell morphology observed via transmission electron microscopy. After establishing the conditions for DHP administration, flow cytometry was employed to assess its effects on apoptosis, reactive oxygen species (ROS), and mitochondrial membrane potential of HCT116 cells. Additionally, immunoprecipitation, quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, and biomarker detection were utilized to investigate the mechanisms underlying DHP's inhibition of HCT116 cells and its impact on metabolic reprogramming. Results In the present study, we observed that DHP treatment at 600 μg/ml for 24 h reduced HCT116 cell viability to 54.87%. In contrast, the inhibitory effect of DHP on the viability of IEC6 and Caco-2 cells was relatively mild. The specific mechanism involves DHP activating the mitochondrial apoptotic pathway leading to the downregulation of key metabolic intermediates and enzymes such as uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) and ST6Gal-I. By inhibiting ST6Gal-I activity, DHP activates the Fas/FasL signaling pathway. Additionally, DHP-induced ROS production effectively triggers apoptosis in HCT116 cells. Conclusion Our study demonstrates that DHP effectively inhibits the proliferation and induces apoptosis in HCT116 colon cancer cells through the activation of the Fas-FasL signaling pathway and metabolic reprogramming. The selective inhibitory effect of DHP on HCT116 cells, the activation of both death receptor and mitochondrial apoptotic pathways, and the modulation of metabolic reprogramming provide novel insights into the potential therapeutic strategies for colon cancer.
Collapse
Affiliation(s)
- Liang Yao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
| | - Chen Gu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
| | - Ruipeng Ge
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
| | - Xiaoqian Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
| | - Xinqian Meng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
| | - Lei Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory for Research and Development of Research and Development of Chinese Medicine, Hefei, China
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory for Research and Development of Research and Development of Chinese Medicine, Hefei, China
| | - Guozhuan Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
| |
Collapse
|
44
|
Severo JS, da Silva ACA, dos Santos BLB, Reinaldo TS, de Oliveira AM, Lima RSP, Torres-Leal FL, dos Santos AA, da Silva MTB. Physical Exercise as a Therapeutic Approach in Gastrointestinal Diseases. J Clin Med 2025; 14:1708. [PMID: 40095789 PMCID: PMC11899784 DOI: 10.3390/jcm14051708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
Background/Objectives: Physical exercise can have significant consequences for the gastrointestinal tract, which is why there have been studies into its influence on the treatment of conditions such as colorectal cancer, inflammatory bowel diseases (IBD), and irritable bowel syndrome (IBS), being that there is epidemiological evidence that exercise has a protective effect against colon cancer. This review aims to demonstrate the mechanisms of action of physical exercise in the gastrointestinal tract, as well as the benefits of exercise in diseases associated with the digestive system, in addition to gathering training recommendations in treating different gastrointestinal diseases. Results: Physical exercise modulates gastrointestinal motility, permeability, immune responses, and microbiota composition, with both beneficial and adverse effects depending on intensity and duration. Regular moderate exercise is associated with improved quality of life in IBD and IBS, reduced colorectal cancer risk, and potential symptom relief in constipation. However, high-intensity exercise may exacerbate gastroesophageal reflux symptoms and increase the risk of gastrointestinal bleeding. While aerobic exercise has been extensively studied, the effects of resistance training on gastrointestinal health remain underexplored. Conclusions: New methodologies and techniques, such as molecular biology and the study of gastric receptors, have led to advances in understanding the gastrointestinal changes associated with physical exercise. These advances cover different exercise intensities and are being investigated in both experimental models and clinical studies.
Collapse
Affiliation(s)
- Juliana Soares Severo
- Graduate Program in Food Sciences and Nutrition, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Laboratory of Metabolic Diseases Glauto Tuquarre, Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piaui, Teresina 64049-550, PI, Brazil; (J.S.S.); (A.M.d.O.); (R.S.P.L.); (F.L.T.-L.)
| | | | | | - Thiago Sousa Reinaldo
- Multicenter Postgraduate Program in Physiological Sciences in Association with the Brazilian Society of Physiology, Federal University of Piauí, Teresina 64049-550, PI, Brazil;
| | - Aureliano Machado de Oliveira
- Graduate Program in Food Sciences and Nutrition, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Laboratory of Metabolic Diseases Glauto Tuquarre, Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piaui, Teresina 64049-550, PI, Brazil; (J.S.S.); (A.M.d.O.); (R.S.P.L.); (F.L.T.-L.)
| | - Rodrigo Soares Pereira Lima
- Graduate Program in Food Sciences and Nutrition, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Laboratory of Metabolic Diseases Glauto Tuquarre, Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piaui, Teresina 64049-550, PI, Brazil; (J.S.S.); (A.M.d.O.); (R.S.P.L.); (F.L.T.-L.)
| | - Francisco Leonardo Torres-Leal
- Graduate Program in Food Sciences and Nutrition, Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Laboratory of Metabolic Diseases Glauto Tuquarre, Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piaui, Teresina 64049-550, PI, Brazil; (J.S.S.); (A.M.d.O.); (R.S.P.L.); (F.L.T.-L.)
| | - Armênio Aguiar dos Santos
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza 60430-270, CE, Brazil;
| | - Moisés Tolentino Bento da Silva
- Graduate Program in Pharmacology, Federal University of Piauí, Teresina 64049-550, PI, Brazil;
- Laboratory of Physiology, (MedInUP/RISE-Health)—Department of Immunophysiology and Pharmacology, School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
45
|
Li Q, Xiao Y, Han L, Luo W, Dai W, Fang H, Wang R, Xu Y, Cai S, Goel A, Bai F, Cai G. Microbiome dysbiosis, neutrophil recruitment and mesenchymal transition of mesothelial cells promotes peritoneal metastasis of colorectal cancer. NATURE CANCER 2025; 6:493-510. [PMID: 39966610 DOI: 10.1038/s43018-025-00910-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/13/2025] [Indexed: 02/20/2025]
Abstract
Peritoneal metastasis (PM) is common in colorectal cancer (CRC), yet its underlying mechanisms are poorly understood. Here, we explored the transcriptional profile of CRC, PM and adjacent tissues revealing key players that facilitate PM. Single-cell analysis of 48 matched samples from 12 patients revealed that remodeling of malignant cells and the tumor microenvironment promotes CRC progression and metastasis. Multiplexed imaging confirmed depletion in PM by enrichment in CRC tissues of neutrophils associated with mucosal immunity disruption, intestinal microbiota dysbiosis and mesenchymal transition of both cancerous and mesothelial cells. Functional analyses in cell lines, organoids and in vivo models demonstrated that dysbiosis promoted inflammation and protumor neutrophil recruitment, while coupled mesenchymal transition of malignant and mesothelial cells disrupted the stromal structure and increased cancer cell invasiveness. Our findings suggest that targeting mesothelial cells and tumor microenvironment remodeling may offer therapeutic strategies for CRC-PM.
Collapse
Affiliation(s)
- Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiwei Xiao
- Biomedical Pioneering Innovation Center (BIOPIC), Peking-Tsinghua Center for Life Sciences (CLS), School of Life Sciences, Peking University, Beijing, China
| | - Lingyu Han
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenqin Luo
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weixing Dai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongsheng Fang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Renjie Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ye Xu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA, USA.
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC), Peking-Tsinghua Center for Life Sciences (CLS), School of Life Sciences, Peking University, Beijing, China.
| | - Guoxiang Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
46
|
Mahmoud MO, Al-Hamid HA, Hassan NF, El-Ansary MR, Gomaa SB. Linagliptin Mitigates DMH-Induced Colorectal Cancer in Rats: Crosstalk Between NFAT and IL-6/JAK2/STAT3/NF-κB Signaling Hubs. J Biochem Mol Toxicol 2025; 39:e70206. [PMID: 40070168 DOI: 10.1002/jbt.70206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 01/30/2025] [Accepted: 03/02/2025] [Indexed: 05/13/2025]
Abstract
Colorectal cancer (CRC) is a multicomponent disease and the second most frequent root of cancer-related deaths globally. Linagliptin is a dipeptidyl peptidase-4 (DPP-4) inhibitor. It has been repurposed in recent experimental studies due to its marked anti-inflammatory activities. This study aimed to evaluate the ameliorative role of linagliptin in 1,2-dimethylhydrazine (DMH)-induced CRC via modulation of NFAT-mediated IL-6 and JAK2/STAT3/NF-κB signaling pathways. CRC model has been successfully established via a dose equal 40 mg/kg two times a week of DMH for 8-week duration. Twenty-four Wistar rats were segregated into three groups of eight rats each; normal control, DMH-induced CRC and DMH + linagliptin (10 mg/kg; p.o). Linagliptin attenuated DMH-induced oxidative stress by restoring the declined levels of some antioxidant enzymes. Linagliptin suppressed the elevated nuclear factor kappa B (NF-κB) induced by DMH which is highlighted using immunohistochemistry analysis. The anti-inflammatory role of linagliptin has been fortified by the decline in nuclear factor of activated T-cells (NFAT) mRNA expression level along with the reduction in vascular endothelial growth factor (VEGF), interlukin-6 (IL-6) and cyclooxygenase-2 (COX-2) levels. Linagliptin mitigate the protein expression of DMH-activated oncogenic janus-activated kinase/signal transducers and activators of transcription (JAK2/STAT3). Linagliptin exerted a proapoptotic effect to tumor cells manifested by a remarkable decline in B-cell lymphoma 2 (Bcl-2) and a significant elevation in Bcl-2-associated X protein (Bax) expression levels. The histopathological analysis revealed that linagliptin has inhibitory potential against the DMH induced dysplastic aberrant crypt foci (ACF) and adenocarcinoma. Linagliptin ameliorated CRC by modulating NFAT-mediated IL-6 with JAK2/STAT3/NF-κB signaling cascades.
Collapse
Affiliation(s)
- Mohamed O Mahmoud
- Biochemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Hager Abd Al-Hamid
- Biochemistry Department, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Noha F Hassan
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Mona R El-Ansary
- Biochemistry Department, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Safaa B Gomaa
- Biochemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
47
|
Duan X, Yeerkenbieke G, Huang S, Feng Y. USP32 Promotes Colorectal Carcinoma Progression Through Activating NF-κB Signalling Pathway. J Cell Mol Med 2025; 29:e70457. [PMID: 40122703 PMCID: PMC11930632 DOI: 10.1111/jcmm.70457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/03/2024] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
Ubiquitin-specific protease 32 (USP32) plays a key role in cancer progression. However, its functions in colorectal carcinoma (CRC) are still unexplored. In our study, we explored the expression and clinical significance of USP32 in CRC as well as its relationship with the tumour microenvironment (TME). As a result, we found that USP32 is overexpressed in CRC and it is associated with poor outcomes in CRC patients. In addition, the expression of USP32 is significantly related to the activation of the NF-κB signalling pathway and the immune infiltrates of the TME. Wet experiments also confirmed that USP32 is critical for the proliferation, survival, and migration of CRC cells and tumour growth, which may be due to the activation of the NF-κB signalling pathway. In conclusion, targeting the USP32-NF-κB axis may be a novel treatment strategy for CRC patients.
Collapse
Affiliation(s)
- Xiaofan Duan
- School of MedicineTongji UniversityShanghaiChina
- Department of OncologyShanghai GoBroad Cancer Hospital, China Pharmaceutical UniversityShanghaiChina
| | - Gaoshaer Yeerkenbieke
- School of MedicineTongji UniversityShanghaiChina
- Department of OncologyShanghai GoBroad Cancer Hospital, China Pharmaceutical UniversityShanghaiChina
| | - Siping Huang
- Department of surgical oncologyShanghai GoBroad Cancer Hospital, China Pharmaceutical UniversityShanghaiChina
| | - Yanjun Feng
- Department of OncologyShanghai GoBroad Cancer Hospital, China Pharmaceutical UniversityShanghaiChina
- Shanghai East Hospital, Nanjing Medical UniversityShanghaiChina
- Department of OncologyShanghai Artemed HospitalShanghaiChina
| |
Collapse
|
48
|
Shahzad M, Hameed H, Amjad A, Khan MA, Qureshi IS, Hameed A, Saeed A, Munir R. An updated landscape on nanopharmaceutical delivery for mitigation of colon cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2107-2125. [PMID: 39361171 DOI: 10.1007/s00210-024-03482-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/21/2024] [Indexed: 03/19/2025]
Abstract
Globally, colorectal cancer (CRC) continues to rank among the leading causes of cancer-related death. Systemic toxicity, multidrug resistance, and nonspecific targeting often pose challenges to conventional therapy for CRC. Because it is a complex disease with a complex genetic and environmental pathophysiology, advanced therapeutic strategies are needed. Nanotechnology presents a potential solution that may maximize therapeutic efficacy while minimizing negative effects by enabling personalized delivery of anticancer drugs. This review focuses on recent developments in colorectal drug delivery systems based on nanotechnology. Numerous nanomaterials, including liposomes, dendrimers, micelles, exosomes, and gold nanoparticles, are developed and used. Distinctive characteristics of mentioned nanocarriers are discussed along with strategies that can be employed for enhancing the delivery of drugs to colorectal cancer cells. The review also quotes the most relevant preclinical and clinical studies that show how these nanomaterials improve drug solubility, stability, and targeted delivery while overcoming the shortcomings of conventional therapies. Nanotechnology has made CRC treatment very efficient and advanced, which has opened up new possibilities for targeted drug delivery. Preclinical and clinical studies have also proved that the use of nano-formulations in colon-specific delivery systems have significant results, indicating potential for better patient outcomes. Future research can be done in order to overcome the hurdles regarding biocompatibility, expansion, and regulatory challenges. Large-scale clinical trials and nanomaterial formulation optimization should be the main goals of future research to confirm the efficacy and safety of these novel treatments.
Collapse
Affiliation(s)
- Maria Shahzad
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Ayesha Amjad
- Faculty of Food Technology and Nutrition Sciences, Lahore University of Biological and Applied Sciences, Lahore, 54000, Pakistan
| | - Mahtab Ahmad Khan
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Inaba Shujaat Qureshi
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore, 54000, Pakistan
| | - Anam Hameed
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore, 54000, Pakistan
| | - Asad Saeed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Rabia Munir
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| |
Collapse
|
49
|
Zhou M, Wang J, Peng Y, Tian X, Zhang W, Chen J, Wang Y, Wang Y, Yang Y, Zhang Y, Huo X, Wu Y, Yu Z, Xie T, Ma X. Elemene as a binding stabilizer of microRNA-145-5p suppresses the growth of non-small cell lung cancer. J Pharm Anal 2025; 15:101118. [PMID: 40161444 PMCID: PMC11953980 DOI: 10.1016/j.jpha.2024.101118] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 04/02/2025] Open
Abstract
Elemene is widely recognized as an effective anti-cancer compound and is routinely administered in Chinese clinical settings for the management of several solid tumors, including non-small cell lung cancer (NSCLC). However, its detailed molecular mechanism has not been adequately demonstrated. In this research, it was demonstrated that elemene effectively curtailed NSCLC growth in the patient-derived xenograft (PDX) model. Mechanistically, employing high-throughput screening techniques and subsequent biochemical validations such as microscale thermophoresis (MST), microRNA-145-5p (miR-145-5p) was pinpointed as a critical target through which elemene exerts its anti-tumor effects. Interestingly, elemene serves as a binding stabilizer for miR-145-5p, demonstrating a strong binding affinity (dissociation constant (K D) = 0.39 ± 0.17 μg/mL) and preventing its degradation both in vitro and in vivo, while not interfering with the synthesis of the primary microRNA transcripts (pri-miRNAs) and precursor miRNAs (pre-miRNAs). The stabilization of miR-145-5p by elemene resulted in an increased level of this miRNA, subsequently suppressing NSCLC progression through the miR-145-5p/mitogen-activated protein kinase kinase kinase 3 (MAP3K3)/nuclear factor kappaB (NF-κB) pathway. Our findings provide a new perspective on revealing the interaction patterns between clinical anti-tumor drugs and miRNAs.
Collapse
Affiliation(s)
- Meirong Zhou
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Jiayue Wang
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Yulin Peng
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Xiangge Tian
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Wen Zhang
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Junlin Chen
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Yue Wang
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Yu Wang
- Research & Production Department, Dalian Huali Jingang Pharmaceutical Co., Ltd., Dalian, Liaoning, 116110, China
| | - Youjian Yang
- Research & Production Department, Dalian Huali Jingang Pharmaceutical Co., Ltd., Dalian, Liaoning, 116110, China
| | - Yongwei Zhang
- Research & Production Department, Dalian Huali Jingang Pharmaceutical Co., Ltd., Dalian, Liaoning, 116110, China
| | - Xiaokui Huo
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Yuzhuo Wu
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Zhenlong Yu
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Tian Xie
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Xiaochi Ma
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
- Shenzhen Bao'an Authentic TCM Therapy Hospital, Shenzhen, Guangdong, 518101, China
| |
Collapse
|
50
|
Zhao M, Zhou L, Zhang Q, Wang M, Dong Y, Wang Y, Pei R, He E, Liang Y, Shen Y, Deng G, Chen H, Sun D, Shen Y, Sun Y, Cheng H. Targeting MAPK14 by Lobeline Upregulates Slurp1-Mediated Inhibition of Alternative Activation of TAM and Retards Colorectal Cancer Growth. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407900. [PMID: 39840525 PMCID: PMC11904982 DOI: 10.1002/advs.202407900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/19/2024] [Indexed: 01/23/2025]
Abstract
Colorectal cancer (CRC) usually creates an immunosuppressive microenvironment, thereby hindering immunotherapy response. Effective treatment options remain elusive. Using scRNA-seq analysis in a tumor-bearing murine model, it is found that lobeline, an alkaloid from the herbal medicine lobelia, promotes polarization of tumor-associated macrophages (TAMs) toward M1-like TAMs while inhibiting their polarization toward M2-like TAMs. Additionally, lobeline upregulates mRNA expression of secreted Ly-6/UPAR-related protein 1 (Slurp1) in cancer cells. The inhibitory effects of lobeline on tumor load and TAM polarization are almost completely eliminated when Slurp1-deficient MC38 cells are subcutaneously injected into mice, suggesting that lobeline exerts an antitumor effect in a Slurp1-dependent manner. Furthermore, using target-responsive accessibility profiling, MAPK14 is identified as the direct target protein of lobeline. Mechanistically, upon binding to MAPK14 in colon cancer cells, lobeline prevents nuclear translocation of MAPK14, resulting in decreased levels of phosphorylated p53. Consequently, negative transcriptional regulation of SLURP1 by p53 is suppressed, leading to enhanced transcription and secretion of SLURP1. Finally, combination therapy using lobeline and anti-PD1 exhibits stronger antitumor effects. Taken together, these findings suggest that remodeling the immunosuppressive microenvironment using small-molecule lobeline may represent a promising therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Mingxia Zhao
- School of Basic Medical SciencesBiopharmaceutical Research InstituteAnhui Medical UniversityHefei230032China
| | - Lisha Zhou
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing Drum Tower Hospitalthe Affiliated Hospital of Nanjing University Medical SchoolSchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
| | - Qinchang Zhang
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of TumorThe First Clinical CollegeNanjing University of Chinese Medicine138 Xianlin AvenueNanjing210023China
| | - Meijing Wang
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing Drum Tower Hospitalthe Affiliated Hospital of Nanjing University Medical SchoolSchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
| | - Yue Dong
- School of Basic Medical SciencesBiopharmaceutical Research InstituteAnhui Medical UniversityHefei230032China
| | - Yue Wang
- School of Basic Medical SciencesBiopharmaceutical Research InstituteAnhui Medical UniversityHefei230032China
| | - Ruixue Pei
- School of Basic Medical SciencesBiopharmaceutical Research InstituteAnhui Medical UniversityHefei230032China
| | - Enguang He
- School of Basic Medical SciencesBiopharmaceutical Research InstituteAnhui Medical UniversityHefei230032China
| | - Yanyan Liang
- School of Basic Medical SciencesBiopharmaceutical Research InstituteAnhui Medical UniversityHefei230032China
| | - Yujun Shen
- School of Basic Medical SciencesBiopharmaceutical Research InstituteAnhui Medical UniversityHefei230032China
| | - Guoliang Deng
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing Drum Tower Hospitalthe Affiliated Hospital of Nanjing University Medical SchoolSchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
| | - Hongqi Chen
- Department of General SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Dongdong Sun
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of TumorThe First Clinical CollegeNanjing University of Chinese Medicine138 Xianlin AvenueNanjing210023China
| | - Yuxian Shen
- School of Basic Medical SciencesBiopharmaceutical Research InstituteAnhui Medical UniversityHefei230032China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing Drum Tower Hospitalthe Affiliated Hospital of Nanjing University Medical SchoolSchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
- Jiangsu Key Laboratory of New Drug Research and Clinical PharmacyXuzhou Medical University209 Tongshan RoadXuzhou221004China
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of TumorThe First Clinical CollegeNanjing University of Chinese Medicine138 Xianlin AvenueNanjing210023China
| |
Collapse
|