1
|
Gan L, Yu CY, Chen J, Zou B, Xiao Z, Jiang W, Li D, Sun Q, Wang Z, Li C, Liu Y, Chu Y, Tang J, Fu M, Li X, Munford R, Lu M. Acyloxyacyl Hydrolase Prevents Colitis and Colitis-Associated Colorectal Cancer by Inactivating Stimulatory LPS in the Intestine. FASEB J 2025; 39:e70566. [PMID: 40277184 DOI: 10.1096/fj.202500310r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/22/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
Ulcerative colitis (UC) is believed to be triggered by a dysregulated inflammatory response to the intestinal microbiota. Acyloxyacyl hydrolase (AOAH) is a unique host lipase that inactivates Gram-negative bacterial lipopolysaccharides (LPS). After finding that AOAH produced in the intestine decreases stimulatory LPS levels in colon contents, we used the dextran sodium sulfate (DSS) model to test the enzyme's ability to prevent colitis in mice. We found that AOAH played a protective role by decreasing colonic inflammation, tissue injury, and barrier permeability. Increasing or decreasing intestinal LPS abundance exacerbated or alleviated colitis, respectively, suggesting that AOAH prevents colitis by reducing stimulatory intestinal LPS levels. AOAH also mitigated colitis-associated colorectal cancer. This highly conserved enzyme may exert its protective effects by preventing LPS-induced injury to the epithelial cell mitochondria that are important for restoring the mucosal epithelial barrier after injury. By decreasing intestinal levels of stimulatory LPS, AOAH prevents colitis and colorectal cancer.
Collapse
Affiliation(s)
- Lu Gan
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Cheng-Yun Yu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jiayi Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Benkun Zou
- BeiGene Institute, BeiGene (Shanghai) Research & Development Co., Ltd, Shanghai, China
| | - Zeling Xiao
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Wei Jiang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Dantong Li
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Qingyang Sun
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Zhiyan Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Changshun Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yiling Liu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Mingsheng Fu
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Xiaobo Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Robert Munford
- Antibacterial Host Defense Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Mingfang Lu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
- MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China
| |
Collapse
|
2
|
Ma Y, Zhou Y, Xie G, Chen H, Huangfu Y, Shen L, Liu Y, Wang P. HEX-1 reduces colitis-driven colorectal cancer via inactivating the prolyl isomerase PIN1 sensitization and remodeling the gut microbiota. Discov Oncol 2025; 16:565. [PMID: 40251462 PMCID: PMC12008109 DOI: 10.1007/s12672-025-02338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
Metabolic reprogramming, a pivotal hallmark of cancer, plays a crucial role in both the initiation and progression of colorectal cancer (CRC). Despite the vast unknowns surrounding the identity and biological activities of most natural metabolites in diseases, our study, utilizing native metabolomics results through GC-MS/MS, identified a small molecule, 4,4-Dimethyl-2-cyclohexen-1-one, named HEX-1 in the serum of CRC patients. We have further explored and assessed its biological activities. HEX-1 suppressed the proliferation of cancer cells and tumorigenesis via the inactivation and sensitization of PIN1. Notably, HEX-1 exhibits similar functional effects as all-trans retinoic acid (atRA) but stands out by not inducing the degradation of PIN1 mRNA or protein expression, unlike biological compounds associated with atRA. HEX-1 demonstrated the ability to induce G1/S arrest in vitro and ameliorate the progression of inflammatory CRC in mice by remodeling the gut microbiota. As volatile organic compounds (VOCs), HEX-1 could be detected feasibly. Its unique ability to penetrate whole cell populations positions it as a promising approach for cancer therapy and as an enhancer for chemotherapy and immunotherapy. The findings suggest that HEX-1 holds the potential as a valuable addition to the armamentarium against CRC.
Collapse
Affiliation(s)
- Yanhui Ma
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yunlan Zhou
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guohua Xie
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Hui Chen
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuchan Huangfu
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Lisong Shen
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, 200092, China
| | - Yi Liu
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, 200092, China.
| | - Ping Wang
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, 200092, China.
| |
Collapse
|
3
|
Janeckova L, Stastna M, Hrckulak D, Berkova L, Kubovciak J, Onhajzer J, Kriz V, Dostalikova S, Mullerova T, Vecerkova K, Tenglerova M, Coufal S, Kostovcikova K, Blumberg RS, Filipp D, Basler K, Valenta T, Kolar M, Korinek V. Tcf4 regulates secretory cell fate decisions in the small intestine and colon tumors: insights from transcriptomic, histological, and microbiome analyses. Stem Cell Res Ther 2025; 16:170. [PMID: 40221753 PMCID: PMC11993999 DOI: 10.1186/s13287-025-04280-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/15/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND The canonical Wnt signaling pathway controls the continuous renewal of the intestinal epithelium and the specification of epithelial cell lineages. Tcf4, a nuclear mediator of Wnt signaling, is essential for the differentiation and maintenance of Paneth cells in the small intestine. Its deficiency is associated with reduced expression of key α-defensins, highlighting its role in host-microbe interactions. However, the exact function of Tcf4 in specifying the secretory lineage and its contribution to antimicrobial peptide production remain incompletely understood. Remarkably, α-defensin expression has also been detected in human colon adenomas, where aberrant Wnt signaling is a hallmark. This raises important questions: What is the role of these Paneth-like cells in tumor biology, and how does Tcf4 influence their identity and function? METHODS We investigated cell specification in small intestinal crypts and colon tumors using conditional Tcf7l2 deletion, cell type-specific Cre recombinases, and reporter alleles in mice. Transcriptomic (single-cell and bulk RNA sequencing) and histological analyses were performed and complemented by microbiome profiling, antibiotic treatment, and intestinal organoids to functionally validate the main findings. RESULTS The inactivation of Tcf4 depletes Paneth cells and antimicrobial peptides, disrupting the gut microbiota balance. In secretory progenitors, loss of Tcf4 shifts differentiation toward goblet cells. In the small intestine, alternative secretory progenitors produce Wnt ligands to support stem cells and epithelial renewal in the absence of Paneth cells. In colon tumors, Paneth-like cells form a tumor cell population, express Wnt ligands, and require Tcf4 for their identity. Loss of Tcf4 redirects their differentiation toward goblet cells. CONCLUSIONS Tcf4 controls the balance between Paneth and goblet cells and is essential for antimicrobial peptide production in the small intestine. In colon adenomas, Paneth-like tumor cells drive antimicrobial gene expression and provide Wnt3 ligands, which may have implications for cancer therapy.
Collapse
Affiliation(s)
- Lucie Janeckova
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Videnska 1083, Prague 4, 142 20, Czech Republic.
| | - Monika Stastna
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Videnska 1083, Prague 4, 142 20, Czech Republic
| | - Dusan Hrckulak
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Videnska 1083, Prague 4, 142 20, Czech Republic
| | - Linda Berkova
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Videnska 1083, Prague 4, 142 20, Czech Republic
| | - Jan Kubovciak
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Jakub Onhajzer
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Videnska 1083, Prague 4, 142 20, Czech Republic
| | - Vitezslav Kriz
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Videnska 1083, Prague 4, 142 20, Czech Republic
| | - Stela Dostalikova
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Videnska 1083, Prague 4, 142 20, Czech Republic
| | - Tereza Mullerova
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Videnska 1083, Prague 4, 142 20, Czech Republic
| | - Katerina Vecerkova
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Marketa Tenglerova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Stepan Coufal
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Klara Kostovcikova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology, Czech Academy of Sciences, Prague, Czech Republic
| | | | - Dominik Filipp
- Laboratory of Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Konrad Basler
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Tomas Valenta
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Videnska 1083, Prague 4, 142 20, Czech Republic
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Michal Kolar
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Vladimir Korinek
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Videnska 1083, Prague 4, 142 20, Czech Republic.
| |
Collapse
|
4
|
Achasova KM, Snytnikova OA, Chanushkina KE, Morozova MV, Tsentalovich YP, Kozhevnikova EN. Baseline abundance of Akkermansia muciniphila and Bacteroides acidifaciens in a healthy state predicts inflammation associated tumorigenesis in the AOM/DSS mouse model. Sci Rep 2025; 15:12241. [PMID: 40210644 PMCID: PMC11985942 DOI: 10.1038/s41598-025-96514-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/28/2025] [Indexed: 04/12/2025] Open
Abstract
Numerous studies demonstrate that intestinal microbiota contribute to colorectal cancer (CRC), which is often associated with dysbiosis. Most of the data were obtained from studies on CRC patients, making it challenging to determine whether alterations in microbiota are a consequence of the pathology or whether they actively drive its progression. Several studies using laboratory animals suggest that gut microbiota may be involved in both the onset and progression of CRC. In the present study we utilized the azoxymethane-dextran sulfate sodium (AOM/DSS) mouse model of CRC to investigate the contribution of healthy-state microbiota to inflammation-associated tumorigenesis. Two cohorts of C57BL/6 mice harboring different intestinal microbiota demonstrated different susceptibility to AOM/DSS treatment. Sequencing of 16S rRNA bacterial DNA from fecal samples revealed Akkermansia muciniphila and Bacteroides acidifaciens as marker features in the healthy-state microbiota (before AOM/DSS administration), which showed a strong positive correlation with tumor incidence. Moreover, the healthy-state abundance of these markers, considered beneficial bacteria, was strongly positively correlated with the sulfate-reducing bacteria Desulfovibrio fairfieldensis identified as a marker of chronic colitis-associated microbiota. Furthermore, the abundances of these marker features, associated with CRC outcome, correlated with the expression of interferon gamma and nitric oxide synthase 2 genes in colon tissue during the early stage of DSS-induced intestinal inflammation. In contrast to multiple studies demonstrating the anti-inflammatory properties of A. muciniphila and B. acidifaciens, our results point out their potential adverse effect under specific conditions of genotoxicity and inflammation in the intestine. Taken together, our findings suggest a complex, context-dependent role of commensal microbiota in inflammation-associated dysbiosis and CRC.
Collapse
Affiliation(s)
- Kseniya M Achasova
- Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia, 630117
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk, Russia, 630090
| | | | | | - Maryana V Morozova
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk, Russia, 630090
| | | | - Elena N Kozhevnikova
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk, Russia, 630090.
- Laboratory of Bioengineering, Novosibirsk State Agrarian University, Novosibirsk, Russia, 630039.
| |
Collapse
|
5
|
Zhou J, Zhang X, Wang C, Xu X, Zhang J, Ge Y, Li J, Yang F, Gao J. An inulin-type fructan CP-A from Codonopsis pilosula combined with 5-Fluorouracil alleviates colitis-associated tumorigenesis via inhibition of EGFR/AKT/ERK signaling pathway and regulation of intestinal flora. Int J Biol Macromol 2025; 308:142655. [PMID: 40158564 DOI: 10.1016/j.ijbiomac.2025.142655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/09/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Inulin-type fructan CP-A, the main component of Codonopsis pilosula polysaccharides, has been found to have therapeutic effects on ulcerative colitis (UC). Herein, we established a colitis-associated cancer (CAC) mouse model by azomethane (AOM) and dextran sulfate sodium (DSS) and selected mouse colon cancer cells CT-26 to explore the therapeutic effects of the combined administration of CP-A and 5-fluorouracil (5-FU) in vivo and in vitro. High-throughput transcriptomics sequencing technology was used to identify differentially expressed genes (DEGs) in the mouse colon and enrich related pathways. 16S rRNA gene sequencing technology was used for gut microbiota research to identify microbial changes in mouse feces. Short-chain fatty acid (SCFA) content was identified in the mouse colon using gas chromatography-mass spectroscopy (GC-MS). In vivo experiments showed that compared with untreated CAC mice, those treated with the combined administration of CP-A and 5-FU had significantly restored body weight, fewer tumors, smaller tumor volume, and reduced disease activity index (DAI) and histopathological scores. The combination of CP-A and 5-FU increased the anti-inflammatory cytokine interleukin 10 (IL-10) and inhibited the expression of pro-inflammatory cytokines interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), and interferon-gamma (IFN-γ). In vitro experiments indicated that a combination of CP-A and 5-FU promoted the apoptosis of CT-26 cells. The results of transcriptomics studies suggested that the therapeutic effect of the combined administration of CP-A and 5-FU on CAC may be related to the EGFR/AKT/ERK pathway. Both in vivo and in vitro experiments verified the regulatory effect of the combined administration of CP-A and 5-FU on the EGFR/AKT/ERK pathway. Moreover, the intestinal flora experiment manifested that compared with untreated CAC mice, the combined CP-A and 5-FU group had a more stable intestinal microbiota composition, and the combined administration of CP-A and 5-FU increased the abundance of SCFAs. Our experimental findings have demonstrated that the combination of CP-A and 5-FU exhibits promising efficacy in the treatment of CAC, warranting further clinical investigation in the future.
Collapse
Affiliation(s)
- Jiangtao Zhou
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China; Shanxi Engineering Research Center of Characteristic Drug Development, School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Xuepeng Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Changjian Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Xiexin Xu
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Jingwen Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Yuhui Ge
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Jiankuan Li
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China; Shanxi Engineering Research Center of Characteristic Drug Development, School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Fan Yang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China; Shanxi Engineering Research Center of Characteristic Drug Development, School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China.
| | - Jianping Gao
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China; Shanxi Engineering Research Center of Characteristic Drug Development, School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
6
|
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] [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
|
7
|
Pickett JR, Wu Y, Ta HT. VCAM-1 as a common biomarker in inflammatory bowel disease and colorectal cancer: unveiling the dual anti-inflammatory and anti-cancer capacities of anti-VCAM-1 therapies. Cancer Metastasis Rev 2025; 44:40. [PMID: 40095109 PMCID: PMC11913972 DOI: 10.1007/s10555-025-10258-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Vascular cell adhesion molecule (VCAM)-1 has garnered significant research attention due to its potential as a disease biomarker and drug target across several inflammatory pathologies-including atherosclerosis, asthma, rheumatoid arthritis, and inflammatory bowel disease (IBD). The VCAM-1 protein has also been noted for its functional involvement in cancer metastasis and drug resistance to conventional chemotherapeutics. Although the anti-inflammatory and anti-cancer facets of VCAM-1 antagonisation have been examined separately, there is yet to be a review that explicitly addresses the functional interrelationship between these mechanisms. Furthermore, the pleiotropic mechanisms of anti-VCAM-1 therapies may present a useful paradigm for designing drug candidates with synergistic anti-inflammatory and anti-tumorigenic effects. The pathological overlap between inflammatory bowel disease (IBD) and colitis-associated colorectal cancer (CRC) serves as the quintessential disease model to observe this therapeutic duality. This review thereby details the adhesive mechanisms of VCAM-1 in colorectal disease-specifically, driving immune cell infiltration during IBD and tumour cell metastasis in CRC-and posits the potential of this receptor as a common drug target for both diseases. To explore this hypothesis, the current progress of novel VCAM-1-directed drug candidates in experimental models of IBD and CRC is also discussed.
Collapse
Affiliation(s)
- Jessica R Pickett
- School of Environment and Science, Griffith University, Nathan Campus, Brisbane, 4111, QLD, Australia
| | - Yuao Wu
- School of Environment and Science, Griffith University, Nathan Campus, Brisbane, 4111, QLD, Australia
| | - Hang Thu Ta
- School of Environment and Science, Griffith University, Nathan Campus, Brisbane, 4111, QLD, Australia.
| |
Collapse
|
8
|
Kaur G, Tiwari P, Singla S, Panghal A, Jena G. The intervention of NLRP3 inflammasome inhibitor: oridonin against azoxymethane and dextran sulfate sodium-induced colitis-associated colorectal cancer in male BALB/c mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03871-z. [PMID: 40035821 DOI: 10.1007/s00210-025-03871-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/31/2025] [Indexed: 03/06/2025]
Abstract
Colorectal cancer (CRC) ranks third globally in cancer diagnoses. The dysregulation of the NLRP3 inflammasome is prominently linked to several types of cancers. Oridonin, a principal component of Rabdosia rubescens, exhibits inhibitory activity against NLRP3 and is well-recognized for its diverse pharmacological benefits. However, its role in an animal model of colitis-associated colorectal cancer (CACC) remains unexplored. In the present study, the effectiveness of oridonin was investigated against CACC, developed using azoxymethane (AOM), a tumour initiator, and dextran sulphate sodium (DSS), a tumour promoter, in male BALB/c mice. The two-stage murine model of inflammation-associated cancer was established by administering AOM (10 mg/kg b.w.; i.p., once) followed by DSS (2% w/v) in drinking water (3 cycles, 7 days/cycle). Over a span of 10 weeks, the dose-dependent (2.5, 5, and 10 mg/kg, b.w.; i.p.) effects of oridonin were investigated in BALB/c mice. Oridonin significantly alleviated CACC severity, as evidenced by reduced DAI scores and restored body weight. Moreover, it attenuated surrogate markers of inflammation, including myeloperoxidase, nitrite, plasma LPS, TNF-α, IL-1β, and DNA damage. Histopathological examination revealed diminished tumorigenesis and apoptotic cells, corroborated by reduced Ki-67 and TNF-α, along with increased p53 expression in the colon. Following oridonin treatment, IHC/immunofluorescence analyses demonstrated a significantly reduced expression of the components of NLRP3 inflammasome including NLRP3, ASC-1, and caspase-1. Notably, the high dose of oridonin (10 mg/kg) consistently exhibited significant protective effects against CACC by modulating various molecular targets. Present findings confirmed the potential of oridonin in the protection of colitis-associated colorectal cancer, providing valuable insights into its mechanism of action and clinical significance.
Collapse
Affiliation(s)
- Gurpreet Kaur
- Facility of Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, 160062, India
| | - Priyanka Tiwari
- Facility of Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, 160062, India
| | - Shivani Singla
- Facility of Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, 160062, India
| | - Archna Panghal
- Facility of Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, 160062, India
| | - Gopabandhu Jena
- Facility of Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, 160062, India.
| |
Collapse
|
9
|
Sun R, Zhang Y, Zhao X, Tang T, Cao Y, Yang L, Tian Y, Zhang Z, Zhang P, Xu F. Temporal and Spatial Metabolic Shifts Revealing the Transition from Ulcerative Colitis to Colitis-Associated Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412551. [PMID: 39840505 PMCID: PMC11923922 DOI: 10.1002/advs.202412551] [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: 10/08/2024] [Revised: 12/17/2024] [Indexed: 01/23/2025]
Abstract
Patients with ulcerative colitis (UC) have a higher risk of developing colorectal cancer (CRC), however, the metabolic shifts during the UC-to-CRC transition remain elusive. In this study, an AOM-DSS-induced three-stage colitis-associated colorectal cancer (CAC) model is constructed and targeted metabolomics analysis and pathway enrichment are performed, uncovering the metabolic changes in this transition. Spatial metabolic trajectories in the "normal-to-normal adjacent tissue (NAT)-to-tumor" transition, and temporal metabolic trajectories in the "colitis-to-dysplasia-to-carcinoma" transition are identified through K-means clustering of 74 spatially and 77 temporally differential metabolites, respectively. The findings reveal two distinct metabolic profile categories during the inflammation-to-cancer progression: those with consistent changes, either increasing (e.g., kynurenic acid, xanthurenic acid) or decreasing (e.g., long-chain fatty acids, LCFAs), and those enriched at specific disease stages (e.g., serotonin). Further analysis of metabolites with consistent temporal trends identifies eicosapentaenoic acid (EPA) as a key metabolite, potentially exerting anti-inflammatory and anti-cancer effects by inhibiting insulin-like growth factor binding protein 5 (IGFBP5). This study reveals novel metabolic mechanisms underlying the transition from UC to CAC and suggests potential targets to delay the progression.
Collapse
Affiliation(s)
- Ruiqi Sun
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Yuanyuan Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Xian Zhao
- Department of Pharmacy, Drum Tower Hospital, China Pharmaceutical University, Nanjing, 210008, P. R. China
| | - Tian Tang
- School of Pharmacy, Air Force Medical University, Xi'an, 710032, P. R. China
| | - Yuepeng Cao
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, 210009, P. R. China
| | - Liu Yang
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, 210009, P. R. China
| | - Yuan Tian
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Zunjian Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Pei Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Fengguo Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| |
Collapse
|
10
|
Hwang S, Park J, Koo SY, Lee SY, Jo Y, Ryu D, Go H, Lee CW. The ubiquitin ligase Pellino1 targets STAT3 to regulate macrophage-mediated inflammation and tumor development. Nat Commun 2025; 16:1256. [PMID: 39893188 PMCID: PMC11787384 DOI: 10.1038/s41467-025-56440-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 01/19/2025] [Indexed: 02/04/2025] Open
Abstract
Receptor-mediated signaling could be modulated by ubiquitination of pathway intermediates, but the role of such modification in the pathogenesis of inflammation and inflammation-related cancer is lesser known. The ubiquitin ligase Pellino1 has been shown to modulate immune signals by enabling various immune cells to respond to their receptor signals effectively. Here, we show that Pellino1 levels are elevated in patients with colitis, patients with colitis-associated colon cancer (CAC), and murine models of these conditions. In a monocyte-specific Pellino1 knock-out mouse model, we find reduced macrophage migration and activation, leading to attenuated development of colitis and CAC in male mice. Mechanistically, Pellino1 targets STAT3 for lysine 63-mediated ubiquitination, resulting in pathogenic activation of STAT3 signaling. Taken together, our findings reveal a macrophage-specific ubiquitination signaling axis in colitis and CAC development and suggest that Pellino1 is a potential candidate for treating chronic inflammation and inflammation-related cancer.
Collapse
Affiliation(s)
- Soeun Hwang
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Junhee Park
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Seo-Young Koo
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Si-Yeon Lee
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Yunju Jo
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, South Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, South Korea
| | - Heounjeong Go
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, South Korea
| | - Chang-Woo Lee
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea.
- Research Institute, Curogen Technology, Suwon, 16419, South Korea.
| |
Collapse
|
11
|
Kim J, Jeon YJ, Chang IY, Lee JH, You HJ. Disruption of the β-catenin destruction complex via Ephexin1-Axin1 interaction promotes colorectal cancer proliferation. Exp Mol Med 2025; 57:151-166. [PMID: 39741188 PMCID: PMC11799323 DOI: 10.1038/s12276-024-01381-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/16/2024] [Accepted: 10/20/2024] [Indexed: 01/02/2025] Open
Abstract
Wnt signaling is essential for cell growth and tumor formation and is abnormally activated in colorectal cancer (CRC), contributing to tumor progression; however, the specific role and regulatory mechanisms involved in tumor development remain unclear. Here, we show that Ephexin1, a guanine nucleotide exchange factor, is significantly overexpressed in CRC and is correlated with increased Wnt/β-catenin pathway activity. Through comprehensive analysis, including RNA sequencing data from TCGA and functional assays, we observed that Ephexin1 promotes tumor proliferation and migration by activating the Wnt/β-catenin pathway. This effect was mediated by the interaction of Ephexin1 with Axin1, a critical component of the β-catenin destruction complex, which in turn enhanced the stability and activity of β-catenin in signaling pathways critical for tumor development. Importantly, our findings also suggest that targeting Ephexin1 may increase the efficacy of Wnt/β-catenin pathway inhibitors in CRC treatment. These findings highlight the potential of targeting Ephexin1 as a strategy for developing effective treatments for CRC, suggesting a novel and promising approach to therapy aimed at inhibiting cancer progression.
Collapse
Affiliation(s)
- Jeeho Kim
- Laboratory of Genomic Instability and Cancer Therapeutics, Gwangju, South Korea
- Department of Pharmacology, Gwangju, South Korea
| | | | | | - Jung-Hee Lee
- Laboratory of Genomic Instability and Cancer Therapeutics, Gwangju, South Korea.
- Department of Cellular and Molecular Medicine, Chosun University School of Medicine, 375 Seosuk-dong, Gwangju, 501-759, South Korea.
| | - Ho Jin You
- Laboratory of Genomic Instability and Cancer Therapeutics, Gwangju, South Korea.
- Department of Pharmacology, Gwangju, South Korea.
| |
Collapse
|
12
|
Li M, Chen H, Yang X, Zhang W, Ma C, Wang Q, Wang X, Gao R. Conditional knockout of the NSD2 gene in mouse intestinal epithelial cells inhibits colorectal cancer progression. Animal Model Exp Med 2025; 8:322-331. [PMID: 38400589 PMCID: PMC11871126 DOI: 10.1002/ame2.12392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 01/16/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Nuclear receptor-binding SET domain 2 (NSD2) is a histone methyltransferase, that catalyzes dimethylation of lysine 36 of histone 3 (H3K36me2) and is associated with active transcription of a series of genes. NSD2 is overexpressed in multiple types of solid human tumors and has been proven to be related to unfavorable prognosis in several types of tumors. METHODS We established a mouse model in which the NSD2 gene was conditionally knocked out in intestinal epithelial cells. We used azoxymethane and dextran sodium sulfate to chemically induce murine colorectal cancer. The development of colorectal tumors were investigated using post-necropsy quantification, immunohistochemistry, and enzyme-linked immunosorbent assay (ELISA). RESULTS Compared with wild-type (WT) control mice, NSD2fl/fl-Vil1-Cre mice exhibited significantly decreased tumor numbers, histopathological changes, and cytokine expression in colorectal tumors. CONCLUSIONS Conditional knockout of NSD2 in intestinal epithelial cells significantly inhibits colorectal cancer progression.
Collapse
Affiliation(s)
- Mengyuan Li
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Hanxue Chen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Xingjiu Yang
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Wenlong Zhang
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Chengyan Ma
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Qinghong Wang
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Xinpei Wang
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Ran Gao
- National Human Diseases Animal Model Resource CenterInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| |
Collapse
|
13
|
Xu H, Wang Y, Liu G, Zhu Z, Shahbazi M, Reis RL, Kundu SC, Shi X, Zu M, Xiao B. Nano-Armed Limosilactobacillus reuteri for Enhanced Photo-Immunotherapy and Microbiota Tryptophan Metabolism against Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410011. [PMID: 39739630 PMCID: PMC11831460 DOI: 10.1002/advs.202410011] [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: 08/21/2024] [Revised: 12/10/2024] [Indexed: 01/02/2025]
Abstract
Despite being a groundbreaking approach to treating colorectal cancer (CRC), the efficacy of immunotherapy is significantly compromised by the immunosuppressive tumor microenvironment and dysbiotic intestinal microbiota. Here, leveraging the superior carrying capacity and innate immunity-stimulating property of living bacteria, a nanomedicine-engineered bacterium, LR-S-CD/CpG@LNP, with optical responsiveness, immune-stimulating activity, and the ability to regulate microbiota metabolome is developed. Immunoadjuvant (CpG) and carbon dot (CD) co-loaded plant lipid nanoparticles (CD/CpG@LNPs) are constructed and conjugated to the surface of Limosilactobacillus reuteri (LR) via reactive oxygen species (ROS)-responsive linkers. The inherent photothermal and photodynamic properties of oral CD/CpG@LNPs induce in situ cytotoxic ROS generation and immunogenic cell death of colorectal tumor cells. The generated neoantigens and the released CpG function as a potent in situ vaccine that stimulates the maturation of immature dendritic cells. The mature dendritic cells and metabolites secreted by LR subsequently facilitated the tumor infiltration of cytotoxic T lymphocytes to eradicate colorectal tumors. The further in vivo results demonstrate that the photo-immunotherapy and intestinal microbial metabolite regulation of LR-S-CD/CpG@LNPs collectively suppressed the growth of orthotopic colorectal tumors and their liver metastases, presenting a promising avenue for synergistic treatment of CRC via the oral route.
Collapse
Affiliation(s)
- Haiting Xu
- State Key Laboratory of Resource InsectsCollege of SericultureTextile, and Biomass SciencesSouthwest UniversityChongqing400715China
| | - Yajun Wang
- Department of PharmacyPersonalized Drug Therapy Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and TechnologyChengdu610054China
| | - Ga Liu
- State Key Laboratory of Resource InsectsCollege of SericultureTextile, and Biomass SciencesSouthwest UniversityChongqing400715China
| | - Zhenhua Zhu
- Department of GastroenterologyThe First Affiliated Hospital of Nanchang UniversityNanchang330006China
| | - Mohammad‐Ali Shahbazi
- Department of Biomedical EngineeringUniversity Medical Center GroningenUniversity of GroningenAntonius Deusinglaan 1Groningen9713 AVNetherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials ScienceUniversity of GroningenAntonius Deusinglaan 1Groningen9713 AVNetherlands
| | - Rui L. Reis
- 3Bs Research GroupI3Bs — Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAvePark, BarcoGuimarães4805‐017Portugal
- ICVS/3B's‐PT Government Associate LaboratoryBragaGuimarães4800‐058Portugal
| | - Subhas C. Kundu
- 3Bs Research GroupI3Bs — Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAvePark, BarcoGuimarães4805‐017Portugal
- ICVS/3B's‐PT Government Associate LaboratoryBragaGuimarães4800‐058Portugal
| | - Xiaoxiao Shi
- State Key Laboratory of Resource InsectsCollege of SericultureTextile, and Biomass SciencesSouthwest UniversityChongqing400715China
| | - Menghang Zu
- State Key Laboratory of Resource InsectsCollege of SericultureTextile, and Biomass SciencesSouthwest UniversityChongqing400715China
| | - Bo Xiao
- Department of PharmacyPersonalized Drug Therapy Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and TechnologyChengdu610054China
| |
Collapse
|
14
|
Tang X, He M, Ren Y, Ji M, Yan X, Zeng W, Lv Y, Li Y, He Y. Traditional Chinese Medicine formulas-based interventions on colorectal carcinoma prevention: The efficacies, mechanisms and advantages. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:119008. [PMID: 39471879 DOI: 10.1016/j.jep.2024.119008] [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: 07/30/2024] [Revised: 10/08/2024] [Accepted: 10/26/2024] [Indexed: 11/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Traditional Chinese Medicine Formulas (TCMFs) represent a distinctive medical approach to disease treatment and have been utilized in clinical practice for treating intestinal diseases for thousands of years. Recently, TCMFs have received increasing attention due to their advantages of high efficiency, safety, as well as low toxicity, providing promising strategies for preventing colorectal carcinoma (CRC). Nonetheless, the potential mechanism of TCMFs in preventing CRC has not been fully elucidated. AIM OF THE STUDY The literature from the past three years was reviewed to highlight the therapeutic effects and underlying mechanisms of TCMFs in preventing CRC. MATERIALS AND METHODS The keywords have been searched, including "traditional Chinese medicine formulas," "herb pairs," "Herbal plant-derived nanoparticles," et al. in "PubMed" and "China National Knowledge Infrastructure (CNKI)," and screened published articles related to the treatment of intestinal precancerous lesions. This review primarily examined the effectiveness and mechanisms of TCMFs in treating intestinal precancerous lesions, highlighting their significant potential in preventing CRC. RESULTS Gegen Qinlian decoction, Shaoyao decoction, Wu Wei Wan, etc., exert substantial therapeutic effects on intestinal precancerous lesions. These therapeutic effects are demonstrated by a reduction in disease activity index scores, suppression of intestinal inflammation, and preservation of body weight and intestinal function, all of which contribute to the effective prevention of CRC. Besides, the classic Chinese herbal pairs and the extracellular vesicle-like nanoparticles of herbaceous plants have demonstrated superior efficacy in the treatment of intestinal precancerous lesions. Mechanistically, protecting the epithelial barrier, regulating gut microbiota as well as related metabolism, modulating macrophage polarization, and maintaining immune balance contribute to the role of TCMFs in CRC prevention. CONCLUSIONS This review demonstrates the great potential and mechanism of TCMFs in CRC prevention and provides a scientific basis for their utilization in CRC prevention.
Collapse
Affiliation(s)
- Xiaojuan Tang
- School of biomedical sciences, Hunan University, Changsha, 410012, Hunan, China; Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha, 410006, Hunan, China; Hunan Academy of Chinese Medicine, Changsha, 410006, Hunan, China.
| | - Min He
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yuan Ren
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Meng Ji
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Xiaoqi Yan
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha, 410006, Hunan, China
| | - Wen Zeng
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yuan Lv
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha, 410006, Hunan, China; Hunan Academy of Chinese Medicine, Changsha, 410006, Hunan, China
| | - Yongmin Li
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha, 410006, Hunan, China; Hunan Academy of Chinese Medicine, Changsha, 410006, Hunan, China
| | - Yongheng He
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha, 410006, Hunan, China; Hunan Academy of Chinese Medicine, Changsha, 410006, Hunan, China; Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
15
|
Yamamoto-Furusho JK, Gutierrez-Herrera FD. Molecular Mechanisms and Clinical Aspects of Colitis-Associated Cancer in Ulcerative Colitis. Cells 2025; 14:162. [PMID: 39936954 PMCID: PMC11817687 DOI: 10.3390/cells14030162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Inflammatory bowel diseases have long been recognized as entities with a higher risk of colorectal cancer. An increasing amount of information has been published regarding ulcerative colitis-associated colorectal cancer and its unique mechanisms in recent decades, as ulcerative colitis constitutes a chronic process characterized by cycles of activity and remission of unpredictable durations and intensities; cumulative genomic alterations occur during active disease and mucosal healing, resulting in a special sequence of events different to the events associated with sporadic colorectal cancer. The recognition of the core differences between sporadic colorectal cancer and colitis-associated cancer is of great importance to understand and guide the directions in which new research could be performed, and how it could be applied to current clinical scenarios. A DSS/AOM murine model has allowed for a better understanding of the pathogenic mechanisms in colitis-associated cancer, as it is currently the closest model to this unique scenario. In this review, we provide a summary of the main molecular mechanisms and the clinical aspects of colitis-associated cancer in ulcerative colitis.
Collapse
Affiliation(s)
- Jesus K. Yamamoto-Furusho
- Inflammatory Bowel Disease Clinic, Department of Gastroenterology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Ciudad de México 14080, Mexico;
| | | |
Collapse
|
16
|
Tian S, Goand UK, Paudel D, Le GV, Tiwari AK, Prabhu KS, Singh V. Processed Dietary Fiber Partially Hydrolyzed Guar Gum Increases Susceptibility to Colitis and Colon Tumorigenesis in Mice. RESEARCH SQUARE 2024:rs.3.rs-5522559. [PMID: 39711544 PMCID: PMC11661293 DOI: 10.21203/rs.3.rs-5522559/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The vital role of naturally occurring dietary fibers (DFs) in maintaining intestinal health has fueled the incorporation of isolated DFs into processed foods. A select group of soluble DFs, such as partially hydrolyzed guar gum (Phgg), are being promoted as dietary supplements to meet recommended DF intake. However, the potential effects of regular consumption of these processed DFs on gastrointestinal health remain largely unknown. The present study assessed the impact of Phgg on the development of intestinal inflammation and colitis-associated colon carcinogenesis (CAC). Wild-type C57BL/6 mice were fed isocaloric diets containing either 7.5% Phgg and 2.5% cellulose (Phgg group) or 10% cellulose (control) for four weeks. To induce colitis, a subgroup of mice from each group was switched to 1.4% dextran sulfate sodium (DSS) in drinking water for seven days. CAC was induced in another subgroup through a single dose of azoxymethane (AOM, 7.5 mg/kg i.p.) followed by three DSS/water cycles. To our surprise, Phgg feeding exacerbated DSS-induced colitis, as evidenced by body weight loss, disrupted colonic crypt architecture, and increased pro-inflammatory markers accompanied by a decrease in anti-inflammatory markers. Additionally, Phgg feeding led to increased colonic expression of genes promoting cell proliferation. Accordingly, extensive colon tumorigenesis was observed in Phgg-fed mice in the AOM/DSS model, whereas the control group exhibited no visible tumors. To investigate whether reducing Phgg has a distinct effect on colitis and CAC development, mice were fed a low-Phgg diet (2.5% Phgg). The low-Phgg group also exhibited increased colitis and tumorigenesis compared to the control, although the severity was markedly lower than in the regular Phgg (7.5%) group, suggesting a dose-dependent effect of Phgg in colitis and CAC development. Our study reveals that Phgg supplementation exacerbates colitis and promotes colon tumorigenesis, warranting further investigation into the potential gastrointestinal health risks associated with processed Phgg consumption.
Collapse
|
17
|
Li T, Luo C, Liu Z, Li J, Han M, Zhang R, Chen Y, Deng H. Nicotinamide mononucleotide protects STAT1 from oxidative stress-induced degradation to prevent colorectal tumorigenesis. MedComm (Beijing) 2024; 5:e70006. [PMID: 39575303 PMCID: PMC11581775 DOI: 10.1002/mco2.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 11/24/2024] Open
Abstract
Colitis, accompanied by the accumulation of reactive oxygen species (ROS) in the intestinal tract, is a risk factor for colorectal cancer (CRC). Our previous studies indicate that nicotinamide mononucleotide (NMN) replenishment reduces chronic inflammation. In this study, we confirm that NMN supplementation reduces inflammatory cytokine levels and oxidative tissue damage in an azoxymethane/dextran sulfate sodium (AOM/DSS)-induced colitis-associated cancer (CAC) model. Mice treated with NMN developed fewer colon tumors than untreated animals under the same AOM/DSS treatment conditions. Quantitative proteomic analysis revealed a decrease in signal transducer and activator of transcription 1 (STAT1) expression in the CAC model. We demonstrate that STAT1 overexpression induces G1 arrest by downregulating CDK6 expression and suppressing tumor cell proliferation and migration. Of note, H2O2 induced trioxidation of the STAT1 protein and promoted its degradation, which was partially reversed by NMN supplementation. Upon H2O2 treatment, Cys155 in STAT1 was oxidized to sulfonic acid, whereas the mutation of Cys155 to alanine abolished ROS-mediated STAT1 degradation. These results indicate that oxidative stress induces STAT1 degradation in tumor cells and possibly in CAC tissues, whereas supplementation with NMN protects STAT1 from oxidation-induced degradation and prevents tumorigenesis. This study provides experimental evidence for the development of NMN-mediated chemoprevention strategies for CRC.
Collapse
Affiliation(s)
- Ting Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingChina
- School of Life Science and TechnologyWuhan Polytechnic UniversityWuhanChina
| | - Chengting Luo
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingChina
- School of Life ScienceYunnan UniversityYunnanChina
| | - Zongyuan Liu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingChina
| | - Jinyu Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingChina
| | - Meng Han
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingChina
| | - Ran Zhang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingChina
| | - Yuling Chen
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingChina
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingChina
| |
Collapse
|
18
|
Holm M, Stepanauskaitė L, Bäckström A, Birgersson M, Socciarelli F, Archer A, Stadler C, Williams C. Spatial profiling of the mouse colonic immune landscape associated with colitis and sex. Commun Biol 2024; 7:1595. [PMID: 39613949 DOI: 10.1038/s42003-024-07276-1] [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: 03/14/2024] [Accepted: 11/15/2024] [Indexed: 12/01/2024] Open
Abstract
Inflammatory intestinal conditions are a major disease burden. Numerous factors shape the distribution of immune cells in the colon, but a spatial characterization of the homeostatic and inflamed colonic immune microenvironment is lacking. Here, we use the COMET platform for multiplex immunofluorescence to profile the infiltration of nine immune cell populations in mice of both sexes (N = 16) with full spatial context, including in regions of squamous metaplasia. Unsupervised clustering, neighborhood analysis, and manual quantification along the proximal-distal axis characterized the colonic immune landscape, quantified cell-cell interactions, and revealed sex differences. The distal colon was the most affected region during colitis, which was pronounced in males, who exhibited a sex-dependent increase of B cells and reduction of M2-like macrophages. Regions of squamous metaplasia exhibited strong infiltration of numerous immune cell populations, especially in males. Females exhibited more helper T cells and neutrophils at homeostasis and increased M2-like macrophage infiltration in the mid-colon upon colitis. Sex differences were corroborated by plasma cytokine profiles. Our results provide a foundation for future studies of inflammatory intestinal conditions.
Collapse
Affiliation(s)
- Matilda Holm
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Lina Stepanauskaitė
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Anna Bäckström
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Madeleine Birgersson
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Fabio Socciarelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Amena Archer
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Charlotte Stadler
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Cecilia Williams
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden.
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
19
|
Chen J, Liu S, Zhang X, Dai X, Li Y, Han Y, Li L. Bondarzewia dickinsii Against Colitis-Associated Cancer Through the Suppression of the PI3K/AKT/COX-2 Pathway and Inhibition of PGE2 Production in Mice. Nutrients 2024; 16:4048. [PMID: 39683442 DOI: 10.3390/nu16234048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Bondarzewia dickinsii (BD) is a newly discovered edible mushroom with rich nutritional components. This study presents a thorough analysis of the components of BD, examining its inhibitory effects and the underlying mechanisms by which BD influences colitis-associated cancer (CAC). METHODS AOM/DSS-induced CAC mice (male C57BL/6) were used, and a histopathological analysis, intestinal microbiota assessment, and metabolomics profiling were carried out, as well as an evaluation of relevant proteins and factors, to investigate the CAC-inhibitory effects of BD. RESULTS BD is rich in nutritional components, including a total sugar content of 37.29% and total protein content of 24.9%. BD significantly diminished colon inflammation, as well as the size and quantity of tumors. In addition, BD modified the diversity of intestinal microbiota and changed the levels of 19 serum metabolites, including arachidonic acid. BD significantly reduced prostaglandin E2 (PGE2) and cyclooxygenase-2 (COX-2) in colon tissue. Furthermore, it was found to inhibit the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/COX-2 signaling pathway. CONCLUSIONS In general, BD inhibited the onset and progression of CAC by modulating the composition of intestinal microbiota and metabolite levels, suppressing the PI3K/AKT/COX-2 pathway, and decreasing PGE2 expression. This study provides a significant reference for the development of BD as a dietary supplement and pharmaceutical agent in the treatment of CAC.
Collapse
Affiliation(s)
- Junliang Chen
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Science and Research Center for Edible Fungi of Qingyuan County, Qingyuan 323800, China
| | - Shuai Liu
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Xin Zhang
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Xiaojing Dai
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Yu Li
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin 300308, China
| | - Yonglin Han
- Science Popularization Service Center of Jilin Province, Changchun 130021, China
| | - Lanzhou Li
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| |
Collapse
|
20
|
Wang D, Zhu L, Liu H, Feng X, Zhang C, Li T, Liu B, Liu L, Sun J, Chang H, Chen S, Guo S, Yang W. Huangqin tang alleviates colitis-associated colorectal cancer via amino acids homeostasisand PI3K/AKT/mtor pathway modulation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118597. [PMID: 39034016 DOI: 10.1016/j.jep.2024.118597] [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: 02/19/2024] [Revised: 04/29/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangqin Tang (HQT), a traditional Chinese medicine formula, is commonly used in clinical practice for the treatment of inflammatory bowel diseases. It has been reported that HQT exerts antitumor effects on colitis-associated colorectal cancer (CAC). However, the mechanism by which HQT interferes with the inflammation-to-cancer transformation remains unclear. AIMS OF THE STUDY The purpose of this study was to dynamically evaluate the efficacy of HQT in alleviating or delaying CAC and to reveal the underlying mechanism. METHODS We established a mouse model of CAC using azoxymethane combined with 1.5% dextran sodium sulphate. The efficacy of HQT was evaluated based on pathological sections and serum biochemical indices. Subsequently, amino acids (AAs) metabolism analyses were performed using ultra-performance liquid chromatography-tandem mass spectrometry, and the phosphatidylinositol 3 kinase/protein kinase B/mechanistic target of rapamycin (PI3K/AKT/mTOR) pathway was detected by western blotting. RESULTS The data demonstrated that HQT could alleviate the development of CAC in the animal model. HQT effectively reduced the inflammatory response, particularly interleukin-6 (IL-6), in the inflammation induction stage, as well as in the stages of proliferation initiation and tumorigenesis. During the proliferation initiation and tumorigenesis stages, immunohistochemistry staining showed that the expression of the proliferation marker Ki67 was reduced, while apoptosis was increased in the HQT group. Accordingly, HQT substantially decreased the levels of specific AAs in the colon with CAC, including glutamic acid, glutamine, arginine, and isoleucine. Furthermore, HQT significantly inhibited the activated PI3K/AKT/mTOR pathway, which may contribute to suppression of cell proliferation and enhancement of apoptosis. CONCLUSION HQT is effective in alleviating and delaying the colon "inflammation-to-cancer". The mechanism of action may involve HQT maintained AAs metabolism homeostasis and regulated PI3K/AKT/mTOR pathway, so as to maintain the balance between proliferation and apoptosis, and then interfere in the occurrence and development of CAC.
Collapse
Affiliation(s)
- Dunfang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Lin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Haifan Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Xue Feng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Caijuan Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Tao Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Bin Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Jingwei Sun
- Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Hao Chang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Siyuan Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Shanshan Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
21
|
Xu Z, Zhao G, Zhang L, Qiao C, Wang H, Wei H, Liu R, Liu P, Zhang Y, Zhu W, You W. Tong-Xie-Yao-Fang induces mitophagy in colonic epithelial cells to inhibit colitis-associated colorectal cancer. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118541. [PMID: 38992403 DOI: 10.1016/j.jep.2024.118541] [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: 01/02/2024] [Revised: 06/10/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Based on the core pathogenesis of hepatosplenic disorder and qi transformation disorder in ulcerative colitis, Tong-Xie-Yao-Fang (TXYF) is a classical traditional Chinese medicine commonly used to treat ulcerative colitis. Our study revealed that it has the potential to prevent colitis-associated colorectal cancer, which embodies the academic concept in traditional Chinese medicine of treating the disease before it develops. AIM OF THE STUDY This study was aimed at evaluating the therapeutic role of TXYF in treating colitis-associated colorectal cancer and exploring its possible underlying mechanisms. MATERIALS AND METHODS A colitis-associated colorectal cancer model was established in mice using azoxymethane and dextran sulfate sodium salt to examine the therapeutic effect of TXYF. The mouse body weights were observed. Hematoxylin-eosin staining was used to evaluate mouse colon histopathology. Colon cancer cells and colon epithelial cells were used to explore the potential molecular mechanisms. The proliferation and apoptosis of cells were detected by CCK8 and cell colony assays, flow cytometry and western blotting. The epithelial-mesenchymal transition (EMT) and mitophagy markers were examined by immunohistochemistry, western blotting, quantitative real-time PCR and immunofluorescence staining. RESULTS TXYF inhibited the tumorigenesis of mice with colitis-associated colorectal cancer and the growth of inflammatory colon cells. TXYF induced mitophagy in colon cancer cells through the PTEN-induced putative kinase 1 (PINK1)/Parkin pathway to reverse EMT, which was consistent with the results in mice with colitis-associated colorectal cancer. CONCLUSIONS The results of the present study demonstrated that TXYF effectively inhibited the progression of colitis-associated colorectal cancer through the PINK1/Parkin pathway, which provides new evidence for prevention strategies for this disease.
Collapse
Affiliation(s)
- Zitong Xu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Gang Zhao
- Department of Anorectal, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Lize Zhang
- Department of Anorectal, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Cuixia Qiao
- Department of Anorectal, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Hao Wang
- Department of President's Office, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Hongyun Wei
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Ruiqing Liu
- Department of Gastroenterological Surgery, The Affiliated Hospital of Qingdao University, Shandong, 266000, China.
| | - Penglin Liu
- Department of Anorectal, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| | - Yuejuan Zhang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Shandong, 266000, China.
| | - Wei Zhu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Shandong, 266000, China.
| | - Wenli You
- Department of Anorectal, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| |
Collapse
|
22
|
Li M, Chen X, Qu P, Shao Z, Shi L, Quan H, Zhao X, Xu J, Shi L, Chen S, Zheng J, Pan ZQ, Bai J. FBXO22 inhibits colitis and colorectal carcinogenesis by regulating the degradation of the S2448-phosphorylated form of mTOR. Proc Natl Acad Sci U S A 2024; 121:e2402035121. [PMID: 39485803 PMCID: PMC11551398 DOI: 10.1073/pnas.2402035121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 09/25/2024] [Indexed: 11/03/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a considerable threat to human health with a significant risk for colorectal cancer (CRC). However, currently, both the molecular pathogenesis and therapeutic treatment of IBD remain limited. In this report, using both systemic and intestinal epithelium-specific gene knockout mouse models, we demonstrate that FBXO22, a substrate receptor within the SKP1-Cullin 1-F-box family of E3 ubiquitin ligases, plays an inhibitory role in the Azoxymethane/Dextran Sodium Sulfate-induced colorectal inflammatory responses and CRC. FBXO22 targets the serine 2448-phosphorylated form of mammalian mechanistic target of rapamycin (pS2448-mTOR) for ubiquitin-dependent degradation. This proteolytic targeting effect is established based on multiple lines of evidence including the results of colon tissue immunoblots, analysis of cultured cells with altered abundance of FBXO22 by depletion or overexpression, comparison of protein decay rate, effects on mTOR substrates S6K1 and 4E-BP1, analysis of protein-protein interactions, phosphor-peptide binding and competition, as well as reconstituted and cellular ubiquitination. Finally, we have shown that mTOR inhibitor rapamycin (RAPA) was able to alleviate the effects of fbxo22 deletion on colorectal inflammatory response and CRC. These RAPA effects are correlated with the ability of RAPA to inhibit pS2448-mTOR, pS6K1, and p4E-BP1. Collectively, our data support a suppressive role for FBXO22 in colorectal inflammation signaling and CRC initiation by targeting pS2448-mTOR for degradation.
Collapse
Affiliation(s)
- Minle Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu221002, China
| | - Xuan Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu221002, China
| | - Pengfei Qu
- Department of Gastroenterology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu221002, China
| | - Zhiying Shao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu221002, China
| | - Lei Shi
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu221002, China
| | - Haoyu Quan
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu221002, China
| | - Xue Zhao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu221002, China
| | - Jian Xu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu221002, China
| | - Luling Shi
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu221002, China
| | - Silu Chen
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing211166, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu221002, China
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu221002, China
| | - Zhen-Qiang Pan
- Department of Oncological Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY10029-6574
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu221002, China
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu221002, China
| |
Collapse
|
23
|
Murmu N, Ghosh P, Namani A, Patra T. Glyoxylate supplementation ameliorates colitis associated colon cancer progression. J Cell Physiol 2024; 239:e31394. [PMID: 39238268 DOI: 10.1002/jcp.31394] [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: 04/05/2024] [Revised: 06/22/2024] [Accepted: 07/19/2024] [Indexed: 09/07/2024]
Abstract
Colon cancer is on the rise in younger adults. Despite multimodal treatment strategies, clinical outcomes in advanced stage colon cancer patients remain poor. Neoadjuvant/adjuvant chemotherapy efficacy is limited due to chemoresistance, toxicity, and negative side effects. Overwhelming evidence supporting the small-molecule metabolites derived from breakdown of food or microbial sources confer an extensive array of host benefits, including chemo-preventive role in colon cancer. Our previous study indicated that the introduction of glyoxylate (Glx), an intermediate product of microbial or plant metabolism, exerts a cytotoxic effect in colon cancer cells. This study was designed to evaluate the effects of Glx on colon cancer with molecular insights. For this, we established an AOM/DSS-induced colitis associated colon cancer model in mice. Supplementation of Glx in vivo reduced colitis associated tumor growth and altered the metabolic characteristics of tumor tissue in mice without initiating any severe liver or renal toxicity. More specifically, intake of glyoxylate accumulated glycine in the colon tissue by elevation of alanine-glyoxylate transferase (AGXT) activity. Glycine accumulation increased intracellular Ca2+ concentration via glycine receptor activation and dysregulation of Ca2+ homeostasis lead to induction of apoptosis that resulted in arresting tumor growth. Interestingly, elevation of AGXT activity or Glx related specific metabolic pathway provides better survival in colon cancer patients. Collectively, our exclusive findings support the exploration of Glx either as a preventive molecule or its inclusion in the treatment regimens for colon cancer.
Collapse
Affiliation(s)
- Nabendu Murmu
- Department of Signal Transduction & Biogenic Amine, Chittaranjan National Cancer Institute, Kolkata, India
| | - Paramita Ghosh
- Department of Signal Transduction & Biogenic Amine, Chittaranjan National Cancer Institute, Kolkata, India
- Department of Biotechnology, Brainware University, Kolkata, India
| | - Akhileshwar Namani
- Department of Molecular Research, Sri Shankara Cancer Hospital and Research Centre, Bangalore, India
| | - Tapas Patra
- Department of Molecular Research, Sri Shankara Cancer Hospital and Research Centre, Bangalore, India
| |
Collapse
|
24
|
Wang D, Zhu L, Liu H, Feng X, Zhang C, Liu B, Li T, Liu L, Chang H, Sun J, Yang L, Yang W. Altered gut metabolites and metabolic reprogramming involved in the pathogenesis of colitis-associated colorectal cancer and the transition of colon "inflammation to cancer". J Pharm Biomed Anal 2024; 253:116553. [PMID: 39486392 DOI: 10.1016/j.jpba.2024.116553] [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: 07/03/2024] [Revised: 10/27/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Colitis-associated colorectal cancer (CAC) is fatal and can develop spontaneously or as a complication of inflammatory bowel diseases. Although co-administration of azoxymethane/dextran sulfate sodium (AOM/DSS) is a classic method for CAC modeling, its limitations need to be addressed. Accordingly, we aimed to optimize the AOM/DSS model to study CAC extensively and further investigate its pathogenic mechanisms relative to microbiota and metabolism. We optimized the CAC model via a single or enhanced injection of AOM combined with different administration modes and varying DSS concentrations. Subsequently, the fecal-microbiota composition was examined using 16S RNA sequencing, and fecal-colon-metabolome profiles were evaluated via ultra-high performance liquid chromatography-mass spectrometry. Two interval injections of AOM combined with 1.5 % DSS-free drinking resulted in a high tumor formation rate, uniform tumor formation, and low mortality. Based on this model, we innovatively divided the pathogenesis of CAC into three stages, namely inflammation induction, proliferation initiation, and tumorigenesis, and examined the pathological characteristics in each stage. Gut microbial dysbiosis and metabolic alteration drove colorectal tumorigenesis by aggravating inflammation while promoting cell proliferation and carcinogenesis in mice. For the first time, we dynamically demonstrated the process of colon "inflammation to cancer" transformation and provided novel insights to clarify the role of amino acid metabolism in the formation of CAC.
Collapse
Affiliation(s)
- Dunfang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Lin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Haifan Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xue Feng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Caijuan Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Bin Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Tao Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hao Chang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jingwei Sun
- Beijing University of Chinese Medicine, Beijing 100700, China
| | - Lei Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
25
|
Au KM, Wilson JE, Ting JPY, Wang AZ. An injectable subcutaneous colon-specific immune niche for the treatment of ulcerative colitis. Nat Biomed Eng 2024; 8:1243-1265. [PMID: 38049469 DOI: 10.1038/s41551-023-01136-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 10/14/2023] [Indexed: 12/06/2023]
Abstract
As a chronic autoinflammatory condition, ulcerative colitis is often managed via systemic immunosuppressants. Here we show, in three mouse models of established ulcerative colitis, that a subcutaneously injected colon-specific immunosuppressive niche consisting of colon epithelial cells, decellularized colon extracellular matrix and nanofibres functionalized with programmed death-ligand 1, CD86, a peptide mimic of transforming growth factor-beta 1, and the immunosuppressive small-molecule leflunomide, induced intestinal immunotolerance and reduced inflammation in the animals' lower gastrointestinal tract. The bioengineered colon-specific niche triggered autoreactive T cell anergy and polarized pro-inflammatory macrophages via multiple immunosuppressive pathways, and prevented the infiltration of immune cells into the colon's lamina propria, promoting the recovery of epithelial damage. The bioengineered niche also prevented colitis-associated colorectal cancer and eliminated immune-related colitis triggered by kinase inhibitors and immune checkpoint blockade.
Collapse
Affiliation(s)
- Kin Man Au
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Justin E Wilson
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jenny P-Y Ting
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew Z Wang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
26
|
Zhang N, Zhang R, Jiang L, Gao Z, Xia W, Ma X, Qin Y, Zhang D, Li J, Tian P, Zhang Q, Wang W, Zhang K, Xu S, Zhao N, Xu S. Inhibition of colorectal cancer in Alzheimer's disease is mediated by gut microbiota via induction of inflammatory tolerance. Proc Natl Acad Sci U S A 2024; 121:e2314337121. [PMID: 39226363 PMCID: PMC11406296 DOI: 10.1073/pnas.2314337121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 07/24/2024] [Indexed: 09/05/2024] Open
Abstract
Epidemiological studies have revealed an inverse relationship between the incidence of Alzheimer's disease (AD) and various cancers, including colorectal cancer (CRC). We aimed to determine whether the incidence of CRC is reduced in AD-like mice and whether gut microbiota confers resistance to tumorigenesis through inducing inflammatory tolerance using 16S ribosomal RNA gene sequencing and fecal microbiota transplantation (FMT). AD-like mice experienced a significantly decreased incidence of CRC tumorigenesis induced by azoxymethane-dextran sodium sulfate as evidenced by suppressed intestinal inflammation compared with control mice. However, FMT from age-matched control mice reversed the inhibitory effects on the tumorigenesis of CRC and inflammatory response in AD-like mice. The key bacterial genera in gut microbiota, including Prevotella, were increased in both the AD-like mice and in patients with amnestic mild cognitive impairment (aMCI) but were decreased in patients with CRC. Pretreatment with low-dose Prevotella-derived lipopolysaccharides (LPS) induced inflammatory tolerance both in vivo and in vitro and inhibited CRC tumorigenesis in mice. Imbalanced gut microbiota increased intestinal barrier permeability, which facilitated LPS absorption from the gut into the blood, causing cognitive decline in AD-like mice and patients with aMCI. These data reveal that intestinal Prevotella-derived LPS exerts a resistant effect to CRC tumorigenesis via inducing inflammatory tolerance in the presence of AD. These findings provide biological evidence demonstrating the inverse relationship between the incidence of AD and CRC.
Collapse
Affiliation(s)
- Nan Zhang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Rui Zhang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Lei Jiang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Zhaoyu Gao
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Wenzhen Xia
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Xiaoying Ma
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Yushi Qin
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Di Zhang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Jiazheng Li
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Pei Tian
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Qi Zhang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Wanchang Wang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Kaixia Zhang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Shan Xu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Na Zhao
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
| | - Shunjiang Xu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang050031, People’s Republic of China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang050031, People’s Republic of China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang050031, People’s Republic of China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Beijing100730, People’s Republic of China
| |
Collapse
|
27
|
Lv G, Wang Q, Lin L, Ye Q, Li X, Zhou Q, Kong X, Deng H, You F, Chen H, Wu S, Yuan L. mTORC2-driven chromatin cGAS mediates chemoresistance through epigenetic reprogramming in colorectal cancer. Nat Cell Biol 2024; 26:1585-1596. [PMID: 39080411 PMCID: PMC11392818 DOI: 10.1038/s41556-024-01473-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 07/07/2024] [Indexed: 09/14/2024]
Abstract
Cyclic GMP-AMP synthase (cGAS), a cytosolic DNA sensor that initiates a STING-dependent innate immune response, binds tightly to chromatin, where its catalytic activity is inhibited; however, mechanisms underlying cGAS recruitment to chromatin and functions of chromatin-bound cGAS (ccGAS) remain unclear. Here we show that mTORC2-mediated phosphorylation of human cGAS serine 37 promotes its chromatin localization in colorectal cancer cells, regulating cell growth and drug resistance independently of STING. We discovered that ccGAS recruits the SWI/SNF complex at specific chromatin regions, modifying expression of genes linked to glutaminolysis and DNA replication. Although ccGAS depletion inhibited cell growth, it induced chemoresistance to fluorouracil treatment in vitro and in vivo. Moreover, blocking kidney-type glutaminase, a downstream ccGAS target, overcame chemoresistance caused by ccGAS loss. Thus, ccGAS coordinates colorectal cancer plasticity and acquired chemoresistance through epigenetic patterning. Targeting both mTORC2-ccGAS and glutaminase provides a promising strategy to eliminate quiescent resistant cancer cells.
Collapse
Affiliation(s)
- Guoqing Lv
- Institute of Biomedical Sciences, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qian Wang
- Department of Urology, The Third Affiliated Hospital & South China Hospital of Shenzhen University, Shenzhen, China
| | - Lin Lin
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Qiao Ye
- Clinical Medicine Laboratory, Air Force Medical Center, Beijing, China
| | - Xi Li
- Institute of Biomedical Sciences, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qian Zhou
- Department of Computer Science, City University of Hong Kong, Hong Kong, China
| | - Xiangzhen Kong
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongxia Deng
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, China
| | - Fuping You
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hebing Chen
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Song Wu
- Department of Urology, The Third Affiliated Hospital & South China Hospital of Shenzhen University, Shenzhen, China.
| | - Lin Yuan
- Institute of Biomedical Sciences, Peking University Shenzhen Hospital, Shenzhen, China.
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
28
|
Hamamah S, Lobiuc A, Covasa M. Antioxidant Role of Probiotics in Inflammation-Induced Colorectal Cancer. Int J Mol Sci 2024; 25:9026. [PMID: 39201713 PMCID: PMC11354872 DOI: 10.3390/ijms25169026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Colorectal cancer (CRC) continues to be a significant contributor to global morbidity and mortality. Emerging evidence indicates that disturbances in gut microbial composition, the formation of reactive oxygen species (ROS), and the resulting inflammation can lead to DNA damage, driving the pathogenesis and progression of CRC. Notably, bacterial metabolites can either protect against or contribute to oxidative stress by modulating the activity of antioxidant enzymes and influencing signaling pathways that govern ROS-induced inflammation. Additionally, microbiota byproducts, when supplemented through probiotics, can affect tumor microenvironments to enhance treatment efficacy and selectively mediate the ROS-induced destruction of CRC cells. This review aims to discuss the mechanisms by which taxonomical shifts in gut microbiota and related metabolites such as short-chain fatty acids, secondary bile acids, and trimethylamine-N-oxide influence ROS concentrations to safeguard or promote the onset of inflammation-mediated CRC. Additionally, we focus on the role of probiotic species in modulating ROS-mediated signaling pathways that influence both oxidative status and inflammation, such as Nrf2-Keap1, NF-κB, and NLRP3 to mitigate carcinogenesis. Overall, a deeper understanding of the role of gut microbiota on oxidative stress may aid in delaying or preventing the onset of CRC and offer new avenues for adjunct, CRC-specific therapeutic interventions such as cancer immunotherapy.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, CA 92103, USA
| | - Andrei Lobiuc
- Department of Medicine and Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 7200229 Suceava, Romania;
| | - Mihai Covasa
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Medicine and Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 7200229 Suceava, Romania;
| |
Collapse
|
29
|
Foelsch K, Pelczar P, Zierz E, Kondratowicz S, Qi M, Mueller C, Alawi M, Huebener S, Clauditz T, Gagliani N, Huber S, Huebener P. Intestinal Epithelia and Myeloid Immune Cells Shape Colitis Severity and Colorectal Carcinogenesis via High-mobility Group Box Protein 1. J Crohns Colitis 2024; 18:1122-1133. [PMID: 38285546 DOI: 10.1093/ecco-jcc/jjae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/11/2024] [Accepted: 01/27/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND High-mobility group box protein 1 [HMGB1] is a ubiquitous nucleoprotein with immune-regulatory properties following cellular secretion or release in sterile and in infectious inflammation. Stool and serum HMGB1 levels correlate with colitis severity and colorectal cancer [CRC] progression, yet recent reports indicate that HMGB1 mainly operates as an intracellular determinant of enterocyte fate during colitis, and investigations into the roles of HMGB1 in CRC are lacking. METHODS Using mice with conditional HMGB1-knockout in enterocytes [Hmgb1ΔIEC] and myeloid cells [Hmgb1ΔLysM], respectively, we explored functions of HMGB1 in pathogenetically diverse contexts of colitis and colitis-associated CRC. RESULTS HMGB1 is overexpressed in human inflammatory bowel disease and gastrointestinal cancers, and HMGB1 protein localises in enterocytes and stromal cells in colitis and CRC specimens from humans and rodents. As previously described, enterocyte HMGB1 deficiency aggravates severe chemical-induced intestinal injury, but not Citrobacter rodentium or T cell transfer colitis in mice. HMGB1-deficient enterocytes and organoids do not exhibit deviant apoptotic or autophagic activity, altered proliferative or migratory capacity, abnormal intestinal permeability, or aberrant DSS-induced organoid inflammation in vitro. Instead, we observed altered in vivo reprogramming of both intestinal epithelia and infiltrating myeloid cells in Hmgb1ΔIEC early during colitis, suggesting HMGB1-mediated paracrine injury signalling. Hmgb1ΔIEC had higher CRC burden than wild types in the Apc+/min model, whereas inflammatory CRC was attenuated in Hmgb1ΔLysM. Cellular and molecular phenotyping of Hmgb1ΔIEC and Hmgb1ΔLysM cancers indicates context-dependent transcriptional modulation of immune signalling and extracellular matrix remodelling via HMGB1. CONCLUSION Enterocytes and myeloid cells context-dependently regulate host responses to severe colitis and maladaptive intestinal wound healing via HMGB1.
Collapse
Affiliation(s)
- Katharina Foelsch
- Department of Internal Medicine, I. Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Penelope Pelczar
- Department of Internal Medicine, I. Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elisabeth Zierz
- Department of Internal Medicine, I. Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephanie Kondratowicz
- Department of Internal Medicine, I. Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Minyue Qi
- Bioinformatics Core Facility, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Mueller
- Bioinformatics Core Facility, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Core Facility, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sina Huebener
- Department of Internal Medicine, I. Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Gagliani
- Department of Internal Medicine, I. Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- Department of Internal Medicine, I. Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Huebener
- Department of Internal Medicine, I. Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
30
|
Pravoverov K, Fatima I, Barman S, Jühling F, Primeaux M, Baumert TF, Singh AB, Dhawan P. IL-22 regulates MASTL expression in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2024; 327:G123-G139. [PMID: 38771154 PMCID: PMC11687961 DOI: 10.1152/ajpgi.00260.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Microtubule-associated serine-threonine kinase-like (MASTL) has recently been identified as an oncogenic kinase given its overexpression in numerous cancers. Our group has shown that MASTL expression is upregulated in mouse models of sporadic colorectal cancer and colitis-associated cancer (CAC). CAC is one of the most severe complications of chronic inflammatory bowel disease (IBD), but a limited understanding of the mechanisms governing the switch from normal healing to neoplasia in IBD underscores the need for increased research in this area. However, MASTL levels in patients with IBD and its molecular regulation in IBD and CAC have not been studied. This study reveals that MASTL is upregulated by the cytokine interleukin (IL)-22, which promotes proliferation and has important functions in colitis recovery; however, IL-22 can also promote tumorigenesis when chronically elevated. Upon reviewing the publicly available data, we found significantly elevated MASTL and IL-22 levels in the biopsies from patients with late-stage ulcerative colitis compared with controls, and that MASTL upregulation was associated with high IL-22 expression. Our subsequent in vitro studies found that IL-22 increases MASTL expression in intestinal epithelial cell lines, which facilitates IL-22-mediated cell proliferation and downstream survival signaling. Inhibition of AKT activation abrogated IL-22-induced MASTL upregulation. We further found an increased association of carbonic anhydrase IX (CAIX) with MASTL in IL-22-treated cells, which stabilized MASTL expression. Inhibition of CAIX prevented IL-22-induced MASTL expression and cell survival. Overall, we show that IL-22/AKT signaling increases MASTL expression to promote cell survival and proliferation. Furthermore, CAIX associates with and stabilizes MASTL in response to IL-22 stimulation.NEW & NOTEWORTHY MASTL is upregulated in colorectal cancer; however, its role in colitis and colitis-associated cancer is poorly understood. This study is the first to draw a link between MASTL and IL-22, a proinflammatory/intestinal epithelial recovery-promoting cytokine that is also implicated in colon tumorigenesis. We propose that IL-22 increases MASTL protein stability by promoting its association with CAIX potentially via AKT signaling to promote cell survival and proliferation.
Collapse
Affiliation(s)
- Kristina Pravoverov
- Eppley Institute, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Iram Fatima
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Susmita Barman
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Frank Jühling
- Inserm U1110, Université de Strasbourg, Institute for Translational Medicine and Liver Disease (ITM), Strasbourg, France
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, Strasbourg, France
| | - Mark Primeaux
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Thomas F Baumert
- Inserm U1110, Université de Strasbourg, Institute for Translational Medicine and Liver Disease (ITM), Strasbourg, France
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, Strasbourg, France
- IHU Strasbourg and Gastroenterology-Hepatology Service, Strasbourg University Hospitals, Strasbourg, France
- Institut Universitaire de France (IUF), Paris, France
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, United States
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, United States
| |
Collapse
|
31
|
Wang F, Xie L, Tang Y, Deng T. Unraveling Crucial Mitochondria-Related Genes in the Transition from Ulcerative Colitis to Colorectal Cancer. Drug Des Devel Ther 2024; 18:3175-3189. [PMID: 39071816 PMCID: PMC11283795 DOI: 10.2147/dddt.s455098] [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: 01/16/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024] Open
Abstract
Purpose To clarify the significance of mitochondria-related differentially expressed genes (MTDEGs) in UC carcinogenesis through a bioinformatics analysis and provide potential therapeutic targets for patients with UC associated colorectal cancer. Methods Microarray GSE37283 was utilized to investigate differentially expressed genes (DEGs) in UC and UC with neoplasia (UCN). MTDEGs were identified by intersecting DEGs with human mitochondrial genes. Utilizing LASSO and random forest analyses, we identified three crucial genes. Subsequently, using ROC curve to investigate the predictive ability of three key genes. Following, three key genes were confirmed in AOM/DSS mice model by Real-time PCR. Finally, single-sample gene set enrichment analysis (ssGSEA) was employed to explore the correlation between the hub genes and immune cells infiltration in UC carcinogenesis. Results The three identified hub MTDEGs (HMGCS2, MAVS, RDH13) may exhibit significant diagnostic specificity in the transition from UC to UCN. Real-time PCR assay further confirmed that the expressions of HMGCS2 and RDH13 were significantly downregulated in UCN mice than that in UC mice. ssGSEA analysis revealed the hub genes were highly associated with CD56dim natural killer cells. Conclusion RDH13, HMGCS2, and MAVS may become diagnostic indicators and potential biomarkers for UCN. Our research has the potential to enhance our understanding of the mechanisms underlying carcinogenesis in UC.
Collapse
Affiliation(s)
- Fanqi Wang
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, People’s Republic of China
- Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Limin Xie
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, People’s Republic of China
- Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Yuan Tang
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, People’s Republic of China
- Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Tuo Deng
- Clinical Immunology Center, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| |
Collapse
|
32
|
van Baarle L, De Simone V, Schneider L, Santhosh S, Abdurahiman S, Biscu F, Schneider R, Zanoletti L, Siqueira de Mello R, Verbandt S, Hu Z, Stakenborg M, Ke BJ, Stakenborg N, Salvador Laureano R, García-Reyes B, Henn J, Toma M, Vanmechelen M, Boeckxstaens G, De Smet F, Garg AD, Ibiza S, Tejpar S, Wehner S, Matteoli G. IL-1R signaling drives enteric glia-macrophage interactions in colorectal cancer. Nat Commun 2024; 15:6079. [PMID: 39030280 PMCID: PMC11271635 DOI: 10.1038/s41467-024-50438-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Enteric glia have been recently recognized as key components of the colonic tumor microenvironment indicating their potential role in colorectal cancer pathogenesis. Although enteric glia modulate immune responses in other intestinal diseases, their interaction with the colorectal cancer immune cell compartment remains unclear. Through a combination of single-cell and bulk RNA-sequencing, both in murine models and patients, here we find that enteric glia acquire an immunomodulatory phenotype by bi-directional communication with tumor-infiltrating monocytes. The latter direct a reactive enteric glial cell phenotypic and functional switch via glial IL-1R signaling. In turn, tumor glia promote monocyte differentiation towards pro-tumorigenic SPP1+ tumor-associated macrophages by IL-6 release. Enteric glia cell abundancy correlates with worse disease outcomes in preclinical models and colorectal cancer patients. Thereby, our study reveals a neuroimmune interaction between enteric glia and tumor-associated macrophages in the colorectal tumor microenvironment, providing insights into colorectal cancer pathogenesis.
Collapse
Affiliation(s)
- Lies van Baarle
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Veronica De Simone
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Linda Schneider
- Department of Surgery, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Sneha Santhosh
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Saeed Abdurahiman
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Francesca Biscu
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Reiner Schneider
- Department of Surgery, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Lisa Zanoletti
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Renata Siqueira de Mello
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Sara Verbandt
- Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Zedong Hu
- Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Michelle Stakenborg
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Bo-Jun Ke
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Nathalie Stakenborg
- Laboratory for Intestinal Neuro-Immune Interaction, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Raquel Salvador Laureano
- Cell Stress and Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Balbina García-Reyes
- Department of Surgery, University Hospital Bonn, Medical Faculty, Bonn, Germany
- Mildred Scheel School of Oncology, Aachen Bonn Cologne Düsseldorf (MSSO ABCD), University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Jonas Henn
- Department of Surgery, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Marieta Toma
- Department of Pathology, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Maxime Vanmechelen
- Translational Cell and Tissue Research Unit, Department of Imaging & Pathology, Laboratory for Precision Cancer Medicine, KU Leuven, Leuven, Belgium
- Leuven Institute for Single-Cell Omics (LISCO), KU Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Laboratory for Intestinal Neuro-Immune Interaction, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Frederik De Smet
- Translational Cell and Tissue Research Unit, Department of Imaging & Pathology, Laboratory for Precision Cancer Medicine, KU Leuven, Leuven, Belgium
- Leuven Institute for Single-Cell Omics (LISCO), KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Cell Stress and Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Sales Ibiza
- Laboratory of Cell Biology & Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Sabine Tejpar
- Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sven Wehner
- Department of Surgery, University Hospital Bonn, Medical Faculty, Bonn, Germany.
| | - Gianluca Matteoli
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium.
- Leuven Institute for Single-Cell Omics (LISCO), KU Leuven, Leuven, Belgium.
| |
Collapse
|
33
|
Tran DH, Kim D, Kesavan R, Brown H, Dey T, Soflaee MH, Vu HS, Tasdogan A, Guo J, Bezwada D, Al Saad H, Cai F, Solmonson A, Rion H, Chabatya R, Merchant S, Manales NJ, Tcheuyap VT, Mulkey M, Mathews TP, Brugarolas J, Morrison SJ, Zhu H, DeBerardinis RJ, Hoxhaj G. De novo and salvage purine synthesis pathways across tissues and tumors. Cell 2024; 187:3602-3618.e20. [PMID: 38823389 PMCID: PMC11246224 DOI: 10.1016/j.cell.2024.05.011] [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: 04/05/2023] [Revised: 03/16/2024] [Accepted: 05/03/2024] [Indexed: 06/03/2024]
Abstract
Purine nucleotides are vital for RNA and DNA synthesis, signaling, metabolism, and energy homeostasis. To synthesize purines, cells use two principal routes: the de novo and salvage pathways. Traditionally, it is believed that proliferating cells predominantly rely on de novo synthesis, whereas differentiated tissues favor the salvage pathway. Unexpectedly, we find that adenine and inosine are the most effective circulating precursors for supplying purine nucleotides to tissues and tumors, while hypoxanthine is rapidly catabolized and poorly salvaged in vivo. Quantitative metabolic analysis demonstrates comparative contribution from de novo synthesis and salvage pathways in maintaining purine nucleotide pools in tumors. Notably, feeding mice nucleotides accelerates tumor growth, while inhibiting purine salvage slows down tumor progression, revealing a crucial role of the salvage pathway in tumor metabolism. These findings provide fundamental insights into how normal tissues and tumors maintain purine nucleotides and highlight the significance of purine salvage in cancer.
Collapse
Affiliation(s)
- Diem H Tran
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Dohun Kim
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rushendhiran Kesavan
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Harrison Brown
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Trishna Dey
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Mona Hoseini Soflaee
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Hieu S Vu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen & German Cancer Consortium, Partner Site, Essen, Germany
| | - Jason Guo
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Divya Bezwada
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Houssam Al Saad
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Feng Cai
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ashley Solmonson
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Halie Rion
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rawand Chabatya
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Salma Merchant
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Nathan J Manales
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Vanina T Tcheuyap
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Megan Mulkey
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Thomas P Mathews
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sean J Morrison
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Hao Zhu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Gerta Hoxhaj
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
34
|
Wang X, Chen D, Guo M, Ning Y, Geng M, Guo J, Gao J, Zhao D, Zhang Y, Li Q, Li L, Li S, Li Y, Xie X, Zuo X, Li J. Oxytocin Alleviates Colitis and Colitis-Associated Colorectal Tumorigenesis via Noncanonical Fucosylation. RESEARCH (WASHINGTON, D.C.) 2024; 7:0407. [PMID: 38979515 PMCID: PMC11228076 DOI: 10.34133/research.0407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/22/2024] [Indexed: 07/10/2024]
Abstract
Colon cancer is increasing worldwide and is commonly regarded as hormone independent, yet recent reports have implicated sex hormones in its development. Nevertheless, the role of hormones from the hypothalamus-hypophysis axis in colitis-associated colorectal cancer (CAC) remains uncertain. In this study, we observed a significant reduction in the expression of the oxytocin receptor (OXTR) in colon samples from both patient with colitis and patient with CAC. To investigate further, we generated mice with an intestinal-epithelium-cell-specific knockout of OXTR. These mice exhibited markedly increased susceptibility to dextran-sulfate-sodium-induced colitis and dextran sulfate sodium/azoxymethane-induced CAC compared to wild-type mice. Our findings indicate that OXTR depletion impaired the inner mucus of the colon epithelium. Mechanistically, oxytocin was found to regulate Mucin 2 maturation through β1-3-N-acetylglucosaminyltransferase 7 (B3GNT7)-mediated fucosylation. Interestingly, we observed a positive correlation between B3GNT7 expression and OXTR expression in human colitis and CAC colon samples. Moreover, the simultaneous activations of OXTR and fucosylation by l-fucose significantly alleviated tumor burden. Hence, our study unveils oxytocin's promising potential as an affordable and effective therapeutic intervention for individuals affected by colitis and CAC.
Collapse
Affiliation(s)
- Xia Wang
- Department of Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Laboratory of Translational Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, Shandong 250012, China
| | - Dawei Chen
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine,
Shandong University, Jinan, Shandong 250012, China
| | - Mengnan Guo
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine,
Shandong University, Jinan, Shandong 250012, China
| | - Yao Ning
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine,
Shandong University, Jinan, Shandong 250012, China
| | - Mingze Geng
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine,
Shandong University, Jinan, Shandong 250012, China
| | - Jing Guo
- Department of Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Laboratory of Translational Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, Shandong 250012, China
| | - Jiahui Gao
- Department of Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Laboratory of Translational Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, Shandong 250012, China
| | - Dong Zhao
- Department of Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Laboratory of Translational Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, Shandong 250012, China
| | - Yupeng Zhang
- Department of Molecular Plant Biology,
Norwegian Institute of Bioeconomy Research, Ås 1430, Norway
| | - Qianpeng Li
- Department of Hematology,
Weifang People’s Hospital, Weifang, Shandong 261000, China
| | - Lixiang Li
- Department of Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Laboratory of Translational Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, Shandong 250012, China
| | - Shiyang Li
- Department of Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, Shandong 250012, China
- Advanced Medical Research Institute,
Shandong University, Jinan, Shandong 250012, China
| | - Yanqing Li
- Department of Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Laboratory of Translational Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, Shandong 250012, China
| | - Xiaoran Xie
- Department of Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Laboratory of Translational Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, Shandong 250012, China
| | - Xiuli Zuo
- Department of Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Laboratory of Translational Gastroenterology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, Shandong 250012, China
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine,
Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
35
|
Ekstedt N, Jamioł-Milc D, Pieczyńska J. Importance of Gut Microbiota in Patients with Inflammatory Bowel Disease. Nutrients 2024; 16:2092. [PMID: 38999840 PMCID: PMC11242987 DOI: 10.3390/nu16132092] [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: 05/19/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
Inflammatory bowel diseases (IBDs), such as Crohn's disease (CD) and ulcerative colitis (UC), are chronic diseases of the digestive system with a multifactorial and not fully understood etiology. There is research suggesting that they may be initiated by genetic, immunological, and lifestyle factors. In turn, all of these factors play an important role in the modulation of intestinal microflora, and a significant proportion of IBD patients struggle with intestinal dysbiosis, which leads to the conclusion that intestinal microflora disorders may significantly increase the risk of developing IBD. Additionally, in IBD patients, Toll-like receptors (TLRs) produced by intestinal epithelial cells and dendritic cells treat intestinal bacterial antigens as pathogens, which causes a disruption of the immune response, resulting in the development of an inflammatory process. This may result in the occurrence of intestinal dysbiosis, which IBD patients are significantly vulnerable to. In this study, we reviewed scientific studies (in particular, systematic reviews with meta-analyses, being studies with the highest level of evidence) regarding the microflora of patients with IBD vs. the microflora in healthy people, and the use of various strains in IBD therapy.
Collapse
Affiliation(s)
- Natalia Ekstedt
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland;
| | - Dominika Jamioł-Milc
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland;
| | - Joanna Pieczyńska
- Department of Food Science and Dietetics, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland;
| |
Collapse
|
36
|
Rodríguez-Santiago Y, Garay-Canales CA, Nava-Castro KE, Morales-Montor J. Sexual dimorphism in colorectal cancer: molecular mechanisms and treatment strategies. Biol Sex Differ 2024; 15:48. [PMID: 38867310 PMCID: PMC11170921 DOI: 10.1186/s13293-024-00623-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/26/2024] [Indexed: 06/14/2024] Open
Abstract
INTRODUCTION Sexual dimorphism significantly influences cancer incidence and prognosis. Notably, females exhibit a lower risk and favorable prognosis for non-reproductive cancers compared to males, a pattern observable beyond the scope of risk behaviors such as alcohol consumption and smoking. Colorectal cancer, ranking third in global prevalence and second in mortality, disproportionately affects men. Sex steroid hormones, particularly estrogens and androgens, play crucial roles in cancer progression, considering epidemiological in vivo and in vitro, in general estrogens imparting a protective effect in females and androgens correlating with an increasing risk of colorectal cancer development. MAIN BODY The hormonal impact on immune response is mediated by receptor interactions, resulting in heightened inflammation, modulation of NF-kB, and fostering an environment conducive to cancer progression and metastasis. These molecules also influence the enteric nervous system, that is a pivotal in neuromodulator release and intestinal neuron stimulation, also contributes to cancer development, as evidenced by nerve infiltration into tumors. Microbiota diversity further intersects with immune, hormonal, and neural mechanisms, influencing colorectal cancer dynamics. A comprehensive understanding of hormonal influences on colorectal cancer progression, coupled with the complex interplay between immune responses, microbiota diversity and neurotransmitter imbalances, underpins the development of more targeted and effective therapies. CONCLUSIONS Estrogens mitigate colorectal cancer risk by modulating anti-tumor immune responses, enhancing microbial diversity, and curbing the pro-tumor actions of the sympathetic and enteric nervous systems. Conversely, androgens escalate tumor growth by dampening anti-tumor immune activity, reducing microbial diversity, and facilitating the release of tumor-promoting factors by the nervous system. These findings hold significant potential for the strategic purposing of drugs to fine-tune the extensive impacts of sex hormones within the tumor microenvironment, promising advancements in colorectal cancer therapies.
Collapse
Affiliation(s)
- Yair Rodríguez-Santiago
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, Mexico City, 04510, México
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Edificio D, 1er piso, Circuito de Posgrados, Ciudad Universitaria, Ciudad de México, 04510, México
| | - Claudia Angelica Garay-Canales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, Mexico City, 04510, México
| | - Karen Elizabeth Nava-Castro
- Grupo de Biología y Química Atmosféricas, Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Ciudad Universitaria, CDMX, 04510, México
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, Mexico City, 04510, México.
| |
Collapse
|
37
|
Lian S, Liu S, Wu A, Yin L, Li L, Zeng L, Zhao M, Zhang L. Branched-Chain Amino Acid Degradation Pathway was Inactivated in Colorectal Cancer: Results from a Proteomics Study. J Cancer 2024; 15:3724-3737. [PMID: 38911385 PMCID: PMC11190764 DOI: 10.7150/jca.95454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/25/2024] [Indexed: 06/25/2024] Open
Abstract
Background: Colorectal cancer (CRC) ranks third in terms of cancer incidence and fourth in terms of cancer-related deaths worldwide. Identifying potential biomarkers of CRC is crucial for treatment and drug development. Methods: In this study, we established a C57B/6N mouse model of colon carcinogenesis using azoxymethane-dextran sodium sulfate (AOM-DSS) treatment for 14 weeks to identify proteins associated with colon cancer. An isobaric tag for relative and absolute quantitation (iTRAQ)-based proteomic analysis was conducted on the cell membrane components enriched in the colonic mucosa. Additionally, tumor tissues and adjacent normal colon tissues were collected from patients with colon cancer for comparative protein and metabolite analyses. Results: In total, 74 differentially expressed proteins were identified in the tumor tissue samples from AOM/DSS-treated mice compared to both the adjacent tissue samples from AOM/DSS-treated mice and tissue samples from saline-treated control mice. Bioinformatics analysis revealed eight downregulated proteins enriched in the branched-chain amino acids pathway (valine, leucine, and isoleucine degradation). Moreover, these proteins are already known to be associated with the survival rate of patients with cancer. Targeted metabolomics showed increased levels of valine, leucine, and isoleucine in tumor tissues compared to those in adjacent normal tissues in patients with colon cancer. Furthermore, a real-time PCR experiment demonstrated that Aldehyde dehydrogenase, mitochondrial (short protein name ALDH2, gene name Aldh2) and Hydroxyacyl-coenzyme A dehydrogenase, mitochondrial (short protein name HCDH, gene name Hadh) (two genes) in the pathway of branched-chain amino acids) were downregulated in patients with colon cancer (colon tumor tissues vs. their adjacent colon tissues). ALDH2 expression was further validated by western blotting in AOM/DSS-treated mouse model and in clinical samples. Conclusion: This study highlighted the inactivation of the branched-chain amino acid degradation pathway in colon cancer and identified ALDH2 and HCDH as potential biomarkers for diagnosing colon cancer and developing new therapeutic strategies.
Collapse
Affiliation(s)
- Shixian Lian
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Siyuan Liu
- Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Ao Wu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Lin Yin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Lei Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Liyan Zeng
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Mingkun Zhao
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Lijun Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| |
Collapse
|
38
|
Ashkenazi-Preiser H, Reuven O, Uzan-Yulzari A, Komisarov S, Cirkin R, Turjeman S, Even C, Twaik N, Ben-Meir K, Mikula I, Cohen-Daniel L, Meirow Y, Pikarsky E, Louzoun Y, Koren O, Baniyash M. The Cross-talk Between Intestinal Microbiota and MDSCs Fuels Colitis-associated Cancer Development. CANCER RESEARCH COMMUNICATIONS 2024; 4:1063-1081. [PMID: 38506672 PMCID: PMC11017962 DOI: 10.1158/2767-9764.crc-23-0421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/24/2023] [Accepted: 03/15/2024] [Indexed: 03/21/2024]
Abstract
Intestinal chronic inflammation is associated with microbial dysbiosis and accumulation of various immune cells including myeloid-derived suppressor cells (MDSC), which profoundly impact the immune microenvironment, perturb homeostasis and increase the risk to develop colitis-associated colorectal cancer (CAC). However, the specific MDSCs-dysbiotic microbiota interactions and their collective impact on CAC development remain poorly understood. In this study, using a murine model of CAC, we demonstrate that CAC-bearing mice exhibit significantly elevated levels of highly immunosuppressive MDSCs, accompanied by microbiota alterations. Both MDSCs and bacteria that infiltrate the colon tissue and developing tumors can be found in close proximity, suggesting intricate MDSC-microbiota cross-talk within the tumor microenvironment. To investigate this phenomenon, we employed antibiotic treatment to disrupt MDSC-microbiota interactions. This intervention yielded a remarkable reduction in intestinal inflammation, decreased MDSC levels, and alleviated immunosuppression, all of which were associated with a significant reduction in tumor burden. Furthermore, we underscore the causative role of dysbiotic microbiota in the predisposition toward tumor development, highlighting their potential as biomarkers for predicting tumor load. We shed light on the intimate MDSCs-microbiota cross-talk, revealing how bacteria enhance MDSC suppressive features and activities, inhibit their differentiation into mature beneficial myeloid cells, and redirect some toward M2 macrophage phenotype. Collectively, this study uncovers the role of MDSC-bacteria cross-talk in impairing immune responses and promoting tumor growth, providing new insights into potential therapeutic strategies for CAC. SIGNIFICANCE MDSCs-dysbiotic bacteria interactions in the intestine play a crucial role in intensifying immunosuppression within the CAC microenvironment, ultimately facilitating tumor growth, highlighting potential therapeutic targets for improving the treatment outcomes of CAC.
Collapse
Affiliation(s)
- Hadas Ashkenazi-Preiser
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Or Reuven
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | | | - Sharon Komisarov
- Department of mathematics, Bar-Ilan University, Ramat Gan, Israel
| | - Roy Cirkin
- Department of mathematics, Bar-Ilan University, Ramat Gan, Israel
| | - Sondra Turjeman
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Carmel Even
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Nira Twaik
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Kerem Ben-Meir
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Ivan Mikula
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Leonor Cohen-Daniel
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Yaron Meirow
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Eli Pikarsky
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Yoram Louzoun
- Department of mathematics, Bar-Ilan University, Ramat Gan, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Michal Baniyash
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| |
Collapse
|
39
|
Fatima I, Ahmad R, Barman S, Gowrikumar S, Pravoverov K, Primeaux M, Fisher KW, Singh AB, Dhawan P. Albendazole inhibits colon cancer progression and therapy resistance by targeting ubiquitin ligase RNF20. Br J Cancer 2024; 130:1046-1058. [PMID: 38278978 PMCID: PMC10951408 DOI: 10.1038/s41416-023-02570-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/08/2023] [Accepted: 12/21/2023] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND The repurposing of FDA-approved drugs for anti-cancer therapies is appealing due to their established safety profiles and pharmacokinetic properties and can be quickly moved into clinical trials. Cancer progression and resistance to conventional chemotherapy remain the key hurdles in improving the clinical management of colon cancer patients and associated mortality. METHODS High-throughput screening (HTS) was performed using an annotated library of 1,600 FDA-approved drugs to identify drugs with strong anti-CRC properties. The candidate drug exhibiting most promising inhibitory effects in in-vitro studies was tested for its efficacy using in-vivo models of CRC progression and chemoresistance and patient derived organoids (PTDOs). RESULTS Albendazole, an anti-helminth drug, demonstrated the strongest inhibitory effects on the tumorigenic potentials of CRC cells, xenograft tumor growth and organoids from mice. Also, albendazole sensitized the chemoresistant CRC cells to 5-fluorouracil (5-FU) and oxaliplatin suggesting potential to treat chemoresistant CRC. Mechanistically, Albendazole treatment modulated the expression of RNF20, to promote apoptosis in CRC cells by delaying the G2/M phase and suppressing anti-apoptotic-Bcl2 family transcription. CONCLUSIONS Albendazole, an FDA approved drug, carries strong therapeutic potential to treat colon cancers which are aggressive and potentially resistant to conventional chemotherapeutic agents. Our findings also lay the groundwork for further clinical testing.
Collapse
Affiliation(s)
- Iram Fatima
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rizwan Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Susmita Barman
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Saiprasad Gowrikumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kristina Pravoverov
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mark Primeaux
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kurt W Fisher
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
40
|
Wang Y, Gao JZ, Sakaguchi T, Maretzky T, Gurung P, Narayanan NS, Short S, Xiong Y, Kang Z. LRRK2 G2019S Promotes Colon Cancer Potentially via LRRK2-GSDMD Axis-Mediated Gut Inflammation. Cells 2024; 13:565. [PMID: 38607004 PMCID: PMC11011703 DOI: 10.3390/cells13070565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a serine-threonine protein kinase belonging to the ROCO protein family. Within the kinase domain of LRRK2, a point mutation known as LRRK2 G2019S has emerged as the most prevalent variant associated with Parkinson's disease. Recent clinical studies have indicated that G2019S carriers have an elevated risk of cancers, including colon cancer. Despite this observation, the underlying mechanisms linking LRRK2 G2019S to colon cancer remain elusive. In this study, employing a colitis-associated cancer (CAC) model and LRRK2 G2019S knock-in (KI) mouse model, we demonstrate that LRRK2 G2019S promotes the pathogenesis of colon cancer, characterized by increased tumor number and size in KI mice. Furthermore, LRRK2 G2019S enhances intestinal epithelial cell proliferation and inflammation within the tumor microenvironment. Mechanistically, KI mice exhibit heightened susceptibility to DSS-induced colitis, with inhibition of LRRK2 kinase activity ameliorating colitis severity and CAC progression. Our investigation also reveals that LRRK2 G2019S promotes inflammasome activation and exacerbates gut epithelium necrosis in the colitis model. Notably, GSDMD inhibitors attenuate colitis in LRRK2 G2019S KI mice. Taken together, our findings offer experimental evidence indicating that the gain-of-kinase activity in LRRK2 promotes colorectal tumorigenesis, suggesting LRRK2 as a potential therapeutic target in colon cancer patients exhibiting hyper LRRK2 kinase activity.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Joyce Z. Gao
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Taylor Sakaguchi
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Thorsten Maretzky
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Prajwal Gurung
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Nandakumar S. Narayanan
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
- Department of Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Sarah Short
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Yiqin Xiong
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Zizhen Kang
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
41
|
Zhang J, Zhang P, Li S, Yu T, Lai X, He Y. Study on the effect and mechanism of Lacticaseibacillus rhamnosus AFY06 on inflammation-associated colorectal cancer induced by AOM/DSS in mice. Front Microbiol 2024; 15:1382781. [PMID: 38572238 PMCID: PMC10987852 DOI: 10.3389/fmicb.2024.1382781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Introduction Lacticaseibacillus rhamnosus AFY06 (LR-AFY06) is a microorganism isolated from naturally fermented yogurt in Xinjiang, China. Methods In this study, we investigated the effects and mechanisms of LR-AFY06 in a mouse model of inflammation-associated colon cancer. The mouse model was established by azoxymethane/dextran sulfate sodium (AOM/DSS) induction. The tumor number in intestinal tissues was counted, and the histopathological analysis was performed on colon tissues. Enzyme-linked immunosorbent assay and real-time quantitative polymerase chain reaction were performed to measure relevant protein levels in colon tissues. Results LR-AFY06 treatment alleviated weight loss, increased organ index, reduced intestinal tumor incidence, improved histopathological damage, decreased the levels of inflammatory cytokines such as interleukin-6 (IL-6), interleukin-1 beta (IL-1β), tumor necrosis factor alpha (TNF-α), nuclear factor κB (NF-κB), and inducible nitric oxide synthase (iNOS) in the serum and colon tissue, downregulated the mRNA expression of inhibitor of NF-κB beta (IκBβ), p65, p50, p52, B-cell lymphoma-2 (Bcl-2), and B-cell lymphoma-extra large (Bcl-xL) in colon tissues, and increased the mRNA expression of Bid and caspase-8. The high concentration of LR-AFY06 exerted a better effect than the low concentration; however, the effect was slightly inferior to that of aspirin. Moreover, LR-AFY06 mitigated the intestinal inflammatory process and inhibited intestinal tumor development by regulating the NF-κB and apoptosis pathways. Discussion The present study indicates the regulatory potential of LR-AFY06 in inflammation-associated colorectal cancer in mice, providing a valuable basis for further research.
Collapse
Affiliation(s)
- Jing Zhang
- Environmental and Quality Inspection College, Chongqing Chemical Industry Vocational College, Chongqing, China
| | - Piyun Zhang
- Department of Gastroenterology and Hepatology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Sijia Li
- Environmental and Quality Inspection College, Chongqing Chemical Industry Vocational College, Chongqing, China
| | - Ting Yu
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Xiangyu Lai
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing Cancer Institute, Chongqing Cancer Hospital, Chongqing, China
| | - Yongpeng He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing Cancer Institute, Chongqing Cancer Hospital, Chongqing, China
| |
Collapse
|
42
|
Chaudhuri SM, Weinberg SE, Wang D, Yalom LK, Montauti E, Iyer R, Tang AY, Torres Acosta MA, Shen J, Mani NL, Wang S, Liu K, Lu W, Bui TM, Manzanares LD, Dehghani Z, Wai CM, Gao B, Wei J, Yue F, Cui W, Singer BD, Sumagin R, Zhang Y, Fang D. Mediator complex subunit 1 architects a tumorigenic Treg cell program independent of inflammation. Cell Rep Med 2024; 5:101441. [PMID: 38428427 PMCID: PMC10983042 DOI: 10.1016/j.xcrm.2024.101441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/18/2023] [Accepted: 02/01/2024] [Indexed: 03/03/2024]
Abstract
While immunotherapy has revolutionized cancer treatment, its safety has been hampered by immunotherapy-related adverse events. Unexpectedly, we show that Mediator complex subunit 1 (MED1) is required for T regulatory (Treg) cell function specifically in the tumor microenvironment. Treg cell-specific MED1 deletion does not predispose mice to autoimmunity or excessive inflammation. In contrast, MED1 is required for Treg cell promotion of tumor growth because MED1 is required for the terminal differentiation of effector Treg cells in the tumor. Suppression of these terminally differentiated Treg cells is sufficient for eliciting antitumor immunity. Both human and murine Treg cells experience divergent paths of differentiation in tumors and matched tissues with non-malignant inflammation. Collectively, we identify a pathway promoting the differentiation of a Treg cell effector subset specific to tumors and demonstrate that suppression of a subset of Treg cells is sufficient for promoting antitumor immunity in the absence of autoimmune consequences.
Collapse
Affiliation(s)
- Shuvam M Chaudhuri
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Samuel E Weinberg
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Dongmei Wang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lenore K Yalom
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Elena Montauti
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Radhika Iyer
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Amy Y Tang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Manuel A Torres Acosta
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jian Shen
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nikita L Mani
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Shengnan Wang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kun Liu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Weiyuan Lu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Triet M Bui
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Laura D Manzanares
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Zeinab Dehghani
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ching Man Wai
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Beixue Gao
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Juncheng Wei
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Feng Yue
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Weiguo Cui
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Benjamin D Singer
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ronen Sumagin
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yana Zhang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Deyu Fang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
43
|
Ke C, Zhou H, Xia T, Xie X, Jiang B. GTP binding protein 2 maintains the quiescence, self-renewal, and chemoresistance of mouse colorectal cancer stem cells via promoting Wnt signaling activation. Heliyon 2024; 10:e27159. [PMID: 38468952 PMCID: PMC10926081 DOI: 10.1016/j.heliyon.2024.e27159] [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: 04/05/2023] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers and the second most deadly cancer across the globe. Colorectal cancer stem cells (CCSCs) fuel CRC growth, metastasis, relapse, and chemoresistance. A complete understanding of the modulatory mechanisms of CCSC biology is essential for developing efficacious CRC treatment. In the current study, we characterized the expression and function of GTP binding protein 2 (GTPBP2) in a chemical-induced mouse CRC model. We found that GTPBP2 was expressed at a higher level in CD133+CD44+ CCSCs compared with other CRC cells. Using a lentivirus-based Cas9/sgRNA system, GTPBP2 expression was ablated in CRC cells in vitro. GTPBP2 deficiency caused the following effects on CCSCs: 1) Significantly accelerating proliferation and increasing the proportions of cells at G1, S, and G2/M phase; 2) Impairing resistance to 5-Fluorouracil; 3) Weakening self-renewal but not impacting cell migration. In addition, GTPBP2 deficiency remarkably decreased β-catenin expression while increasing β-catenin phosphorylation in CCSCs. These effects of GTPBP2 were present in CCSCs but not in other CRC cell populations. The Wnt agonist SKL2001 completely abolished these changes in GTPBP2-deficient CCSCs. When GTPBP2-deficient CCSCs were implanted in nude mice, they exhibited consistent changes compared with GTPBP2-expressing CCSCs. Collectively, this study indicates that GTPBP2 positively modulates Wnt signaling to reinforce the quiescence, self-renewal, and chemoresistance of mouse CCSCs. Therefore, we disclose a novel mechanism underlying CCSC biology and GTPBP2 could be a therapeutic target in future CRC treatment.
Collapse
Affiliation(s)
- Chao Ke
- The Department of Gastrointestinal, Hernia and Abdominal Wall Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), 241 Pengliuyang Road, Wuchang District, Wuhan, Hubei Province, 430060, China
| | - Hongjian Zhou
- The Department of Gastrointestinal, Hernia and Abdominal Wall Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), 241 Pengliuyang Road, Wuchang District, Wuhan, Hubei Province, 430060, China
| | - Tian Xia
- The Department of Gastrointestinal, Hernia and Abdominal Wall Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), 241 Pengliuyang Road, Wuchang District, Wuhan, Hubei Province, 430060, China
| | - Xingwang Xie
- The Department of Gastrointestinal, Hernia and Abdominal Wall Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), 241 Pengliuyang Road, Wuchang District, Wuhan, Hubei Province, 430060, China
| | - Bin Jiang
- The Department of Gastrointestinal, Hernia and Abdominal Wall Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), 241 Pengliuyang Road, Wuchang District, Wuhan, Hubei Province, 430060, China
| |
Collapse
|
44
|
Eich C, Vogt JF, Längst V, Clausen BE, Hövelmeyer N. Isolation and high-dimensional flow cytometric analysis of tumor-infiltrating leukocytes in a mouse model of colorectal cancer. Front Immunol 2024; 15:1295863. [PMID: 38500875 PMCID: PMC10944955 DOI: 10.3389/fimmu.2024.1295863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 02/09/2024] [Indexed: 03/20/2024] Open
Abstract
Colorectal cancer (CRC) is a complex and heterogeneous disease characterized by dysregulated interactions between tumor cells and the immune system. The tumor microenvironment plays a pivotal role in cancer initiation as well as progression, with myeloid immune cells such as dendritic cell and macrophage subsets playing diverse roles in cancer immunity. On one hand, they exert anti-tumor effects, but they can also contribute to tumor growth. The AOM/DSS colitis-associated cancer mouse model has emerged as a valuable tool to investigate inflammation-driven CRC. To understand the role of different leukocyte populations in tumor development, the preparation of single cell suspensions from tumors has become standard procedure for many types of cancer in recent years. However, in the case of AOM/DSS-induced colorectal tumors, this is still challenging and rarely described. For one, to be able to properly distinguish tumor-associated immune cells, separate processing of cancerous and surrounding colon tissue is essential. In addition, cell yield, due to the low tumor mass, viability, as well as preservation of cell surface epitopes are important for successful flow cytometric profiling of tumor-infiltrating leukocytes. Here we present a fast, simple, and economical step-by-step protocol for isolating colorectal tumor-associated leukocytes from AOM/DSS-treated mice. Furthermore, we demonstrate the feasibility of this protocol for high-dimensional flow cytometric identification of the different tumor-infiltrating leukocyte populations, with a specific focus on myeloid cell subsets.
Collapse
Affiliation(s)
- Christina Eich
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Johannes F. Vogt
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Vivian Längst
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Björn E. Clausen
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Nadine Hövelmeyer
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
45
|
Song J, Ham J, Park W, Song G, Lim W. Osthole impairs mitochondrial metabolism and the autophagic flux in colorectal cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155383. [PMID: 38295666 DOI: 10.1016/j.phymed.2024.155383] [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: 10/17/2023] [Revised: 01/05/2024] [Accepted: 01/20/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Osthole is active constituent of Cnidium monnieri (L.) Cuss. with various physiological functions including anti-inflammation and anti-lipedemic effects. However, the regulatory activity of osthole in colorectal cancer development, focusing on mitochondrial metabolism, is not well known. HYPOTHESIS/PURPOSE We hypothesized that osthole may suppress progression of colorectal cancer and aimed to determine the underlying mitochondrial metabolism and the autophagic flux. STUDY DESIGN In this study, we elucidated the mechanism of action of osthole in colorectal cancer using an in vivo azoxymethane/dextran sodium sulfate (AOM/DSS) mouse model and an in vitro cell culture system. METHODS AOM/DSS mouse model was established and analyzed the effects of osthole on survival rate, diseases activity index, number of tumor and histopathology. Then, cell based assays including viability, cell cycle, reactive oxygen species (ROS), apoptosis, calcium efflux, and mitochondrial function were analyzed. Moreover, osthole-mediated signaling was demonstrated by western blot analyses. RESULTS Osthole effectively suppressed the growth of colorectal tumors and alleviated AOM/DSS-induced intestinal injury. Osthole restored the function of goblet cells and impaired the expression of Claudin1 and Axin1 impaired by AOM/DSS. In addition, osthole specifically showed cytotoxicity in colorectal carcinoma cells, but not in normal colon cells. Osthole decreased the ASC/caspase-1/IL-1β inflammasome pathway and induced mitochondrial dysfunction in redox homeostasis, calcium homeostasis. Furthermore, osthole inhibited both oxidative phosphorylation (OXPHOS) and glycolysis, leading to the suppression of ATP production. Moreover, via combination treatment with chloroquine (CQ), we demonstrated that osthole impaired autophagic flux, leading to apoptosis of HCT116 and HT29 cells. Finally, we elucidated that the functional role of tiRNAHisGTG regulated by osthole directly affects the cellular fate of colon cancer cells. CONCLUSION These results suggest that osthole has the potential to manage progression of colorectal cancer by regulating autophagy- and mitochondria-mediated signal transduction.
Collapse
Affiliation(s)
- Jisoo Song
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jiyeon Ham
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Wonhyoung Park
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
46
|
Wan Z, Zheng G, Zhang Z, Ruan Q, Wu B, Wei G. Material basis and core chemical structure of Dendrobium officinale polysaccharides against colitis-associated cancer based on anti-inflammatory activity. Int J Biol Macromol 2024; 262:130056. [PMID: 38365160 DOI: 10.1016/j.ijbiomac.2024.130056] [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: 11/02/2023] [Revised: 12/26/2023] [Accepted: 02/06/2024] [Indexed: 02/18/2024]
Abstract
It has been claimed that Dendrobium officinale polysaccharides (PSs) can degrade into oligosaccharide and then transform into short-chain fatty acids in the intestine after oral administration, and play an anti-colitis-associated cancer (CAC) effect by inhibiting intestinal inflammation. However, the material basis and core chemical structure underlying the anti-colon cancer properties of PSs have not yet been elucidated. In this study, PSs were degraded into enzymatic oligosaccharides (OSs) using β-mannanase. The results of in vivo experiments revealed that PSs and OSs administered by gastric lavage had similar antitumor effects in CAC mice. OS-1 (Oligosaccharide compounds 1) and OS-2 (Oligosaccharide compounds 2) were further purified and characterized from OSs, and it was found that OS-1, OS-2, OSs, and PSs had similar and consistent anti-inflammatory activities in vitro. Chemical structure comparison and evaluation revealed that the chemical structure of β-D-Manp-(1 → 4)-β-D-Glcp corresponding to OS-1 was the least common PS structure with anti-colitic activity. Therefore, our findings suggest that OSs are the material basis for PSs to exert anti-CAC activity and that the chemical structure of β-D-Manp-(1 → 4)-β-D-Glcp corresponding to OS-1 is the core chemical structure of PSs against CAC.
Collapse
Affiliation(s)
- Zhongxian Wan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232 Wai Huan Dong Road, Higher Education Mega Center, Panyu District, Guangzhou 511400, China; The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No. 158 Wuyang Dadao, Enshi, Hubei 445000, China
| | - Guoyao Zheng
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No. 158 Wuyang Dadao, Enshi, Hubei 445000, China
| | - Zixiong Zhang
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No. 158 Wuyang Dadao, Enshi, Hubei 445000, China
| | - Qingfeng Ruan
- Department of Pharmacy, Wuhan No.1 Hospital, No. 215 Zhongshan Dadao, Qiaokou District, Wuhan, Hubei 430022, China
| | - Bo Wu
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No. 158 Wuyang Dadao, Enshi, Hubei 445000, China.
| | - Gang Wei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232 Wai Huan Dong Road, Higher Education Mega Center, Panyu District, Guangzhou 511400, China.
| |
Collapse
|
47
|
Peng J, Zhou J, Liu X, Zhang X, Zhou X, Gong Z, Chen Y, Shen X, Chen Y. A biomimetic nanocarrier facilitates glucose consumption and reactive oxide species accumulation in enzyme therapy for colorectal cancer. J Control Release 2024; 367:76-92. [PMID: 38262488 DOI: 10.1016/j.jconrel.2024.01.041] [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: 10/13/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/25/2024]
Abstract
Glucose oxidase (GOx)-based enzyme therapeutics are potential alternatives for colorectal cancer (CRC) treatment via glucose consumption and accumulation of hydrogen peroxide (H2O2). Given that H2O2 can be eliminated by cytoprotective autophagy, autophagy inhibitors that can interrupt autolysosome-induced H2O2 elimination are promising combination drugs of GOx. Here, we developed a multifunctional biomimetic nanocarrier for effective co-delivery of an autophagy inhibitor-chloroquine phosphate (CQP) and GOx to exert their synergistic effect by irreversibly upregulating intracellular reactive oxygen species (ROS) levels. Poly (D, l-lactide-co-glycolide) (PLGA) nanoparticles (NPs) were used to encapsulate both GOx and CQP using a W/O/W multi-emulsion method. Calcium phosphate (CaP) was used to "fix" CQP to GOx in the internal water phase, where it served as a pH-sensitive unit to facilitate intracellular drug release. Folic acid-modified red blood cell membranes (FR) were used to camouflage the GOx/CQP/CaP encapsulated PLGA NPs (referred to as PLGA/GCC@FR). In an AOM/DSS-induced CRC mouse model, PLGA/GCC@FR exhibited improved antitumor effects, in which the number of tumor nodes were only a quarter of that in the free drug combination group. The enhanced therapeutic effects of PLGA/GCC@FR were attributed to the prolonged tumor retention which was verified by both dynamic in vivo imaging and drug biodistribution. This multifunctional biomimetic nanocarrier facilitated combined enzyme therapeutics by depleting glucose and augmenting intracellular ROS levels in tumor cells, which exerted a synergistic inhibitory effect on tumor growth. Therefore, this study proposed a novel strategy for the enhancement of combined enzyme therapeutics, which provided a promising method for effective CRC treatment.
Collapse
Affiliation(s)
- Jianqing Peng
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Jia Zhou
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Xing Liu
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Xiaobo Zhang
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Xiang Zhou
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Zipeng Gong
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Yi Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| | - Xiangchun Shen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| | - Yan Chen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| |
Collapse
|
48
|
Sykes DJ, Solanki S, Chukkapalli S, Williams K, Newman EA, Resnicow K, Shah YM. Structural enrichment attenuates colitis-associated colon cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580099. [PMID: 38405737 PMCID: PMC10888747 DOI: 10.1101/2024.02.13.580099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Colorectal cancer (CRC) is a major public health concern and disproportionately impacts racial/ethnic minority populations in the US. Animal models are helpful in examining human health disparities because many stress-induced human health conditions can be recapitulated using mouse models. Azoxymethane (AOM)/ dextran sodium sulfate (DSS) treatment can be used to model colitis-associated cancers. While colitis-associated cancers account for only 2% of colon cancers, the AOM/DSS model is useful for examining links between inflammation, immunity, and colon cancer. Mice were housed in enriched and impoverished environments for 1-month prior to behavioral testing. Following behavioral testing the mice were subjected to the AOM/DSS model. While our analysis revealed no significant behavioral variances between the impoverished and enriched housing conditions, we found significant effects in tumorigenesis. Enriched mice had fewer tumors and smaller tumor volumes compared to impoverished mice. African Americans are at higher risk for early onset colorectal cancers in part due to social economic status. Furthermore, housing conditions and environment may reflect social economic status. Research aimed at understanding links between social economic status and colorectal cancer progression is important for eliminating disparities in health outcomes.
Collapse
|
49
|
Gu LM, Li HZ, Gao L, Li H, Wei LF, Pan CY, Wu KX, Tian YZ. Huangqin Decoction Delays Progress of Colitis-Associated Carcinogenesis by Regulating Nrf2/HO-1 Antioxidant Signal Pathway in Mice. Chin J Integr Med 2024; 30:135-142. [PMID: 37434030 DOI: 10.1007/s11655-023-3554-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2023] [Indexed: 07/13/2023]
Abstract
OBJECTIVE To investigate the effect of Huangqin Decoction (HQD) on nuclear factor erythroid 2 related-factor 2 (Nrf2)/heme oxygenase (HO-1) signaling pathway by inducing the colitis-associated carcinogenesis (CAC) model mice with azoxymethane (AOM)/dextran sodium sulfate (DSS). METHODS The chemical components of HQD were analyzed by liquid chromatography-quadrupole-time-of-flight mass spectrometry (LC-Q-TOF-MS/MS) to determine the molecular constituents of HQD. Totally 48 C57BL/6J mice were randomly divided into 6 groups by a random number table, including control, model (AOM/DSS), mesalazine (MS), low-, medium-, and high-dose HQD (HQD-L, HQD-M, and HQD-H) groups, 8 mice in each group. Except for the control group, the mice in the other groups were intraperitoneally injected with AOM (10 mg/kg) and administrated with 2.5% DSS orally for 1 week every two weeks (totally 3 rounds of DSS) to construct a colitis-associated carcinogenesis mouse model. The mice in the HQD-L, HQD-M and HQD-H groups were given HQD by gavage at doses of 2.925, 5.85, and 11.7 g/kg, respectively; the mice in the MS group was given a suspension of MS at a dose of 0.043 g/kg (totally 11 weeks). The serum levels of malondialdehyde (MDA) and superoxide dismutase (SOD) were measured by enzyme-linked immunosorbent assay. The mRNA and protein expression levels of Nrf2, HO-1, and inhibitory KELCH like ECH-related protein 1 (Keap1) in colon tissue were detected by quantitative real-time PCR, immunohistochemistry, and Western blot, respectively. RESULTS LC-Q-TOF-MS/MS analysis revealed that the chemical constituents of HQD include baicalin, paeoniflorin, and glycyrrhizic acid. Compared to the control group, significantly higher MDA levels and lower SOD levels were observed in the model group (P<0.05), whereas the expressions of Nrf2 and HO-1 were significantly decreased, and the expression of Keap1 increased (P<0.01). Compared with the model group, serum MDA level was decreased and SOD level was increased in the HQD-M, HQD-H and MS groups (P<0.05). Higher expressions of Nrf2 and HO-1 were observed in the HQD groups. CONCLUSION HQD may regulate the expression of Nrf2 and HO-1 in colon tissue, reduce the expression of MDA and increase the expression of SOD in serum, thus delaying the progress of CAC in AOM/DSS mice.
Collapse
Affiliation(s)
- Li-Mei Gu
- Department of Gastrointestinal Endoscopy, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - He-Zhong Li
- Department of Gastroenterology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Lei Gao
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hui Li
- Department of Gastroenterology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Lan-Fu Wei
- Department of Gastroenterology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Cheng-Yu Pan
- Department of Gastroenterology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Ke-Xuan Wu
- Department of Gastroenterology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Yao-Zhou Tian
- Department of Gastroenterology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
| |
Collapse
|
50
|
Wang L, Tu Y, Chen L, Yu K, Wang H, Yang S, Zhang Y, Zhang S, Song S, Xu H, Yin Z, Feng M, Yue J, Huang X, Tang T, Wei S, Liang X, Chen Z. Black rice diet alleviates colorectal cancer development through modulating tryptophan metabolism and activating AHR pathway. IMETA 2024; 3:e165. [PMID: 38868519 PMCID: PMC10989083 DOI: 10.1002/imt2.165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/15/2023] [Indexed: 06/14/2024]
Abstract
Consumption of dietary fiber and anthocyanin has been linked to a lower incidence of colorectal cancer (CRC). This study scrutinizes the potential antitumorigenic attributes of a black rice diet (BRD), abundantly rich in dietary fiber and anthocyanin. Our results demonstrate notable antitumorigenic effects in mice on BRD, indicated by a reduction in both the size and number of intestinal tumors and a consequent extension in life span, compared to control diet-fed counterparts. Furthermore, fecal transplants from BRD-fed mice to germ-free mice led to a decrease in colonic cell proliferation, coupled with maintained integrity of the intestinal barrier. The BRD was associated with significant shifts in gut microbiota composition, specifically an augmentation in probiotic strains Bacteroides uniformis and Lactobacillus. Noteworthy changes in gut metabolites were also documented, including the upregulation of indole-3-lactic acid and indole. These metabolites have been identified to stimulate the intestinal aryl hydrocarbon receptor pathway, inhibiting CRC cell proliferation and colorectal tumorigenesis. In summary, these findings propose that a BRD may modulate the progression of intestinal tumors by fostering protective gut microbiota and metabolite profiles. The study accentuates the potential health advantages of whole-grain foods, emphasizing the potential utility of black rice in promoting health.
Collapse
Affiliation(s)
- Ling Wang
- Hubei Hongshan Laboratory, Hubei Key Laboratory of Agricultural Bioinformatics, Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, College of Life Science and Technology, College of Biomedicine and Health, Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
- Shenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityShenzhenChina
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
- Department of Pharmaceutical ChemistryUniversity of California‐San FranciscoSan FranciscoCaliforniaUSA
| | - Yi‐Xuan Tu
- Hubei Hongshan Laboratory, Hubei Key Laboratory of Agricultural Bioinformatics, Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, College of Life Science and Technology, College of Biomedicine and Health, Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
- Shenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityShenzhenChina
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| | - Lu Chen
- Hubei Hongshan Laboratory, Hubei Key Laboratory of Agricultural Bioinformatics, Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, College of Life Science and Technology, College of Biomedicine and Health, Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
| | - Ke‐Chun Yu
- Hubei Hongshan Laboratory, Hubei Key Laboratory of Agricultural Bioinformatics, Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, College of Life Science and Technology, College of Biomedicine and Health, Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
| | - Hong‐Kai Wang
- Hubei Hongshan Laboratory, Hubei Key Laboratory of Agricultural Bioinformatics, Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, College of Life Science and Technology, College of Biomedicine and Health, Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
| | - Shu‐Qiao Yang
- Hubei Hongshan Laboratory, Hubei Key Laboratory of Agricultural Bioinformatics, Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, College of Life Science and Technology, College of Biomedicine and Health, Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
| | - Yuan Zhang
- Hubei Hongshan Laboratory, Hubei Key Laboratory of Agricultural Bioinformatics, Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, College of Life Science and Technology, College of Biomedicine and Health, Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
| | - Shuai‐Jie Zhang
- Hubei Hongshan Laboratory, Hubei Key Laboratory of Agricultural Bioinformatics, Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, College of Life Science and Technology, College of Biomedicine and Health, Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
| | - Shuo Song
- Hubei Hongshan Laboratory, Hubei Key Laboratory of Agricultural Bioinformatics, Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, College of Life Science and Technology, College of Biomedicine and Health, Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
| | - Hong‐Li Xu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical CollegeHuazhong Agricultural UniversityWuhanChina
| | - Zhu‐Cheng Yin
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical CollegeHuazhong Agricultural UniversityWuhanChina
| | - Ming‐Qian Feng
- Hubei Hongshan Laboratory, Hubei Key Laboratory of Agricultural Bioinformatics, Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, College of Life Science and Technology, College of Biomedicine and Health, Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
| | - Jun‐Qiu Yue
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | | | - Tang Tang
- Wuhan Metware Biotechnology Co., LtdWuhanChina
| | - Shao‐Zhong Wei
- Department of Gastrointestinal Oncology Surgery, Hubei Cancer Hospital, Tongji Medical CollegeHuazhong Agricultural UniversityWuhanChina
| | - Xin‐Jun Liang
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical CollegeHuazhong Agricultural UniversityWuhanChina
| | - Zhen‐Xia Chen
- Hubei Hongshan Laboratory, Hubei Key Laboratory of Agricultural Bioinformatics, Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, College of Life Science and Technology, College of Biomedicine and Health, Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
- Shenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityShenzhenChina
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| |
Collapse
|