1
|
Liu L, Shi J, Wang H, Du H, Yang J, Wei K, Zhou Z, Li M, Huang S, Zhan L, Li G, Lv Y, Shen H, Cai W. The characteristics of tissue microbiota in different anatomical locations and different tissue types of the colorectum in patients with colorectal cancer. mSystems 2025:e0019825. [PMID: 40422085 DOI: 10.1128/msystems.00198-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/27/2025] [Indexed: 05/28/2025] Open
Abstract
The gut microbiota is intricately associated with the onset and progression of colorectal cancer (CRC), leading to significant interest in developing prevention and treatment strategies that leverage gut microbiota. In this study, we collected 57 samples from 19 CRC patients, comprising cancerous tissue, paracancerous tissue, and normal mucosa. Utilizing metagenomic sequencing and bioinformatics analysis, we identified differences in the microbiomes and their functional characteristics across the various tissue types. The results indicated that species such as Alistipes putredinis were predominantly found in normal tissues, while Pseudomonas putida was enriched in paracancerous tissue, and Malassezia restricta was prevalent in cancerous tissues. Furthermore, the microbial functions exhibited variability among the different tissue types. Random forest analysis suggested that Moraxella osloensis may be implicated in the onset and progression of colorectal cancer. We also classified the patients into three subgroups based on the anatomical location of the colorectum: right-sided colon, left-sided colon, and rectum. The subgroup analysis revealed that the microbiota enriched in normal mucosa and paracancerous tissue varied across different anatomical sites. These findings not only elucidate the characteristics of the microbiomes in the normal mucosa, paracancerous tissue, and cancerous tissues of CRC patients, thereby providing new potential targets for clinical diagnosis and treatment, but also contribute to the existing microbiome data pertinent to CRC research.IMPORTANCEThis study provides crucial insights into the relationship between gut microbiota and colorectal cancer (CRC) by analyzing microbial communities in different tissue types and anatomical locations of CRC patients. We identified distinct microbial signatures, such as Alistipes putredinis in normal tissues and Malassezia restricta in cancerous tissues, indicating location-specific microbiomes with unique functional attributes. These findings suggest potential new biomarkers or therapeutic targets for CRC. The observed microbiota variations among right-sided colon, left-sided colon, and rectum cancers underscore the heterogeneity of CRC, pointing toward more personalized treatment strategies. By enhancing our understanding of the microbiome's role in CRC, this research paves the way for innovative diagnostic tools and targeted therapies tailored to individual patient profiles. This work is essential for advancing clinical approaches to CRC management.
Collapse
Affiliation(s)
- Lei Liu
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Jianguo Shi
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Hui Wang
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Hansong Du
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Jia Yang
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kai Wei
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Zhuohui Zhou
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Moli Li
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Shuai Huang
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lifang Zhan
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Guolong Li
- School of Life Sciences, Hubei University, Wuhan, Hubei, China
| | - Yongling Lv
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
- School of Life Sciences, Hubei University, Wuhan, Hubei, China
| | - Hexiao Shen
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
- School of Life Sciences, Hubei University, Wuhan, Hubei, China
| | - Wei Cai
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| |
Collapse
|
2
|
Lahouty M, Fadaee M, Aghaei R, Alizadeh F, Jafari A, Sharifi Y. Gut microbiome and colorectal cancer: From pathogenesis to treatment. Pathol Res Pract 2025; 271:156034. [PMID: 40412026 DOI: 10.1016/j.prp.2025.156034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/06/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Colorectal cancer (CRC) continues to rank among the most prevalent cancers worldwide. A growing body of research indicates that the microbiome significantly influences the onset, development, and progression of CRC, in addition to affecting the efficacy of various systemic therapies. The composition of the microbiome, shaped by factors such as bacterial strains, geography, ethnicity, gender, and dietary habits, provides essential information for CRC screening, early diagnosis, and the prediction of treatment responses. Modulating the microbiome presents a highly promising medical strategy for improving individual health. This review aims to present a thorough overview of recent research concerning the interplay between host microbiota and CRC, along with its implications for screening and the immune response against tumors in the context of cancer treatment.
Collapse
Affiliation(s)
- Masoud Lahouty
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran.
| | - Reza Aghaei
- Department of veterinary medicine, Shab.C, Islamic Azad University, Shabestar, Iran
| | - Fatemeh Alizadeh
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirmohammad Jafari
- Department of veterinary medicine, Shab.C, Islamic Azad University, Shabestar, Iran
| | - Yaeghob Sharifi
- Department of Microbiology and Virology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
3
|
Yincharoen P, Mordmuang A, Techarang T, Tangngamsakul P, Kaewubon P, Atipairin P, Janwanitchasthaporn S, Goodla L, Karnjana K. Microbiome and biofilm insights from normal vs tumor tissues in Thai colorectal cancer patients. NPJ Precis Oncol 2025; 9:98. [PMID: 40185839 PMCID: PMC11971325 DOI: 10.1038/s41698-025-00873-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 03/10/2025] [Indexed: 04/07/2025] Open
Abstract
Colorectal cancer (CRC) is a prevalent global malignancy with complex etiologies, including microbiota alterations. This study investigates gut microbiota and biofilm-producing bacteria in 35 Thai CRC patients, analyzing paired normal and tumor biopsy samples. Bacterial DNA from the V3-V4 region of 16S rRNA was sequenced, and biofilms were visualized via scanning electron microscopy and fluorescence in situ hybridization (FISH). Results revealed Firmicutes as the dominant phylum, followed by Bacteroidota, Proteobacteria, and Fusobacteriota, with Fusobacteriota and Bacteroidota notably enriched in left-sided CRC. Key biofilm producers-Bacteroides fragilis, Fusobacterium nucleatum, and Pasteurella stomatis-showed significantly higher gene expression in tumor tissues. Dense biofilms and higher Fusobacterium abundance, localized within the crypts of Lieberkuhn, were observed in CRC tissues. These findings highlight CRC-associated microbiota alterations and pathogenic biofilm production, emphasizing a spatial relationship between tumor location and microbial distribution, with potential implications for understanding CRC pathogenesis and therapeutic targeting.
Collapse
Affiliation(s)
- Pirada Yincharoen
- Department of Clinical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand
| | - Auemphon Mordmuang
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand
| | - Tachpon Techarang
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Panus Tangngamsakul
- Walailak University Hospital, Walailak University, Nakhon Si Thammarat, Thailand
| | | | - Paijit Atipairin
- Department of Surgery, Thasala Hospital, Nakhon Si Thammarat, Thailand
| | | | - Lavanya Goodla
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| | - Kulwadee Karnjana
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand.
| |
Collapse
|
4
|
Zhou YL, Deng JW, Liu ZH, Ma XY, Zhu CQ, Xie YH, Zhou CB, Fang JY. Derivation and validation of lifestyle-based and microbiota-based models for colorectal adenoma risk evaluation and self-prediction. BMJ Open Gastroenterol 2025; 12:e001597. [PMID: 40175093 PMCID: PMC12001358 DOI: 10.1136/bmjgast-2024-001597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 03/11/2025] [Indexed: 04/04/2025] Open
Abstract
OBJECTIVE Early warning and screening of colorectal adenoma (CRA) is important for colorectal cancer (CRC) prevention. This study aimed to construct a non-invasive prediction model to improve CRA screening efficacy. METHODS This study incorporated three cohorts, comprising 9747 participants who underwent colonoscopy. In cohort 1, 683 participants were prospectively recruited with comprehensive lifestyle information and faecal samples. CRA-associated bacteria were identified through 16S rRNA sequencing and quantitative real-time PCR. CRA prediction models were established using lifestyle and gut microbiota information. Cohort 2 prospectively enrolled 1529 participants to validate the lifestyle-based model, while cohort 3 retrospectively analysed 7535 individuals to determine the recommended initial colonoscopy screening ages for different risk groups based on age-specific CRA incidence rates. RESULTS Multivariable logistic regression yielded a prediction model incorporating 14 variables, demonstrating robust discrimination (c-statistic=0.79, 95% CI 0.75, 0.82). Other machine learning approaches showed comparable performance (random forest: 0.78, 95% CI 0.73, 0.81; gradient boosting: 0.78, 95% CI 0.76, 0.83). The ages for starting colonoscopy screening were established at 42 years for the high-risk group vs 53 years for the low-risk group. The inclusion of Fusobacterium nucleatum and pks+ Escherichia coli enhanced the model's performance (c-statistic=0.84-0.86). CONCLUSION Integrated mathematical modelling incorporating lifestyle parameters and gut microbial signatures provides an effective non-invasive strategy for CRA risk stratification, while the accompanying machine learning-assisted prediction application enables cost-effective, population-level screening implementation to optimise CRC prevention protocols.
Collapse
Affiliation(s)
- Yi-Lu Zhou
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jia-Wen Deng
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhu-Hui Liu
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xin-Yue Ma
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chun-Qi Zhu
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan-Hong Xie
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng-Bei Zhou
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
5
|
Sujaya IN, Mariyatun M, Hasan PN, Manurung NEP, Pramesi PC, Juffrie M, Utami T, Cahyanto MN, Yamamoto S, Takahashi T, Asahara T, Akiyama T, Rahayu ES. Randomized study of Lacticaseibacillus fermented milk in Indonesian elderly houses: Impact on gut microbiota and gut environment. World J Gastroenterol 2025; 31:104081. [PMID: 40182598 PMCID: PMC11962840 DOI: 10.3748/wjg.v31.i12.104081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/21/2025] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Health maintenance in elderly houses includes management of the gut microbiota and the environment. Lacticaseibacillus paracasei Shirota (LcS) is a probiotic strain that positively affects the human gut. However, the evidence of its effects on the Indonesian population remains limited. AIM To investigate the effect of LcS-fermented milk on the gut microbiota and environment of Indonesian elderly houses. METHODS This double-blind, randomized, placebo-controlled trial involved 112 participants from Indonesian elderly houses, spanning a 2-week baseline and 24-week treatment. Participants were randomly assigned to probiotic or placebo groups, consuming fermented milk with or without LcS (> 6.5 × 109 colony-forming units). Fecal samples were collected every three months. Gut microbiota analysis was performed using 16S rRNA gene sequencing and reverse transcription quantitative polymerase chain reaction, while gut environment was assessed by measuring fecal organic acids, amino acid metabolites, and stool frequency. RESULTS Analyses of 16S rRNA gene sequence data at the 3-month period revealed increased Bifidobacterium and Succinivibrio and decreased Rikenellaceae RC9 gut group in the probiotic group. These shifts were associated with significant differences in β-diversity metrics. The change in Bifidobacterium was confirmed by reverse transcription quantitative polymerase chain reaction, demonstrating higher abundance in the probiotic group than in the placebo group (8.5 ± 1.1 vs 8.0 ± 1.1, log10 bacterial cells/g; P = 0.044). At 6-month period, the differences in Succinivibrio and Rikenellaceae RC9 gut group persisted. The probiotic group showed higher butyrate levels than the placebo group at the 6-month period (5.04 ± 3.11 vs 3.95 ± 2.89, μmol/g; P = 0.048). The effect on amino acid metabolites and stool frequency was not significant. CONCLUSION Daily intake of LcS positively affects the gut microbiota and environment of people living in Indonesian elderly houses.
Collapse
Affiliation(s)
- I Nengah Sujaya
- School of Public Health, Faculty of Medicine, Udayana University, Denpasar 80230, Bali, Indonesia
| | - Mariyatun Mariyatun
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Pratama Nur Hasan
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Nancy Eka Putri Manurung
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Putrika Citta Pramesi
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Mohammad Juffrie
- Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Tyas Utami
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Muhammad Nur Cahyanto
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Shuta Yamamoto
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi 186-8650, Tōkyō, Japan
| | - Takuya Takahashi
- Yakult Honsha European Research Center for Microbiology VOF, Ghent 9052, East Flanders, Belgium
| | - Takashi Asahara
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi 186-8650, Tōkyō, Japan
| | - Takuya Akiyama
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi 186-8650, Tōkyō, Japan
| | - Endang Sutriswati Rahayu
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| |
Collapse
|
6
|
Cao Y, Yang Z, Tang Q. The association between oxidative balance score with constipation and diarrhea in US adults: a cross-sectional analysis of NHANES 2007-2010. BMC Gastroenterol 2025; 25:29. [PMID: 39844079 PMCID: PMC11752723 DOI: 10.1186/s12876-025-03597-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Oxidative stress is associated with functional bowel disorders. The Oxidative Balance Score (OBS) is used to represent the overall level of oxidative stress experienced by an individual. Sex differences are exhibited in the occurrence of constipation and diarrhea. This population-based study aimed to examine the association between the OBS, constipation, and diarrhea, among adults in the United States (US), stratified by sex. METHODS Using data from the National Health and Nutrition Examination Survey (NHANES) database spanning from 2007 to 2010, a cross-sectional study including 4,462 participants (2,386 males and 2,076 females) was performed to assess the relationship between the OBS, constipation, and diarrhea, among adult men and women in the US. Constipation and diarrhea were identified according to bowel frequency and stool type, as outlined in the Bowel Health Questionnaire. The independent variable was the OBS based on 20 types of dietary and lifestyle components. We conducted a binary logistic regression subgroup analysis by sex, and a univariate subgroup analysis; and used a restricted cubic spline (RCS) to illustrate the relationship between the OBS and abnormal bowel health. RESULTS After adjusting for all covariates, a multivariate binary logistic regression analysis revealed that the OBS was negatively correlated with constipation in males [odds ratios (OR) (95% confidence intervals (CI)): 0.937 (0.888, 0.988), P = 0.017], and diarrhea in females [OR (95%CI): 0.951 (0.916, 0.987), P = 0.009]. RCS analysis identified a negative linear trend between OBS and constipation in males (P for non-linearity = 0.63), as well as between OBS and diarrhea in females (P for non-linearity > 0.05). CONCLUSION The current study found a negative correlation between the OBS and the risk of constipation in males, along with diarrhea in females. Oxidative stress, gut microbiota, and sex differences may be associated with intestinal dysfunction. These findings suggest that antioxidant-rich dietary modifications may be an effective strategy for preventing constipation in males and diarrhea in females.
Collapse
Affiliation(s)
- Yaoling Cao
- Department of General Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zhihao Yang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Qinqing Tang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China.
| |
Collapse
|
7
|
Jiang Y, Huang Z, Sun W, Huang J, Xu Y, Liao Y, Jin T, Li Q, Ho IHT, Zou Y, Zhu W, Li Q, Qin F, Zhang X, Shi S, Zhang N, Yang S, Xie W, Wu S, Tan L, Zhang L, Chen H, Gin T, Chan MTV, Wu WKK, Xiao L, Liu X. Roseburia intestinalis-derived butyrate alleviates neuropathic pain. Cell Host Microbe 2025; 33:104-118.e7. [PMID: 39706182 DOI: 10.1016/j.chom.2024.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Approximately 20% of patients with shingles develop postherpetic neuralgia (PHN). We investigated the role of gut microbiota in shingle- and PHN-related pain. Patients with shingles or PHN exhibited significant alterations in their gut microbiota with microbial markers predicting PHN development among patients with shingles. Functionally, fecal microbiota transplantation from patients with PHN to mice heightened pain sensitivity. Administration of Roseburia intestinalis, a bacterium both depleted in patients with shingles and PHN, alleviated peripheral nerve injury-induced pain in mice. R. intestinalis enhanced vagal neurotransmission to the nucleus tractus solitarius (NTS) to suppress the central amygdala (CeA), a brain region involved in pain perception. R. intestinalis-generated butyrate activated vagal neurons through the receptor, G protein-coupled receptor 41 (GPR41). Vagal knockout of Gpr41 abolished the effects of R. intestinalis on the NTS-CeA circuit and reduced pain behaviors. Overall, we established a microbiota-based model for PHN risk assessment and identified R. intestinalis as a potential pain-alleviating probiotic.
Collapse
Affiliation(s)
- Yanjun Jiang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ziheng Huang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wuping Sun
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Jiabin Huang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Yunlong Xu
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Neonatology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen 518055, China
| | - Yuliang Liao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Tingting Jin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qing Li
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Idy Hiu Ting Ho
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yidan Zou
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wenyi Zhu
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qian Li
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fenfen Qin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xinyi Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shuqi Shi
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Na Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shaomin Yang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Wenhui Xie
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Songbin Wu
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Likai Tan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lin Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Huarong Chen
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tony Gin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Matthew Tak Vai Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - William Ka Kei Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; State Key Laboratory of Digestive Disease Institute of Digestive Disease and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Lizu Xiao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China.
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
8
|
Abdullah, Ahmad N, Xiao J, Tian W, Khan NU, Hussain M, Ahsan HM, Hamed YS, Zhong H, Guan R. Gingerols: Preparation, encapsulation, and bioactivities focusing gut microbiome modulation and attenuation of disease symptoms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156352. [PMID: 39740381 DOI: 10.1016/j.phymed.2024.156352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 10/17/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Gut dysbiosis, chronic diseases, and microbial recurrent infections concerns have driven the researchers to explore phytochemicals from medicinal and food homologous plants to modulate gut microbiota, mitigate diseases, and inhibit pathogens. Gingerols have attracted attention as therapeutic agents due to their diverse biological activities like gut microbiome regulation, gastro-protective, anti-inflammatory, anti-microbial, and anti-oxidative effects. PURPOSE This review aimed to summarize the gingerols health-promoting potential, specifically focusing on the regulation of gut microbiome, attenuation of disease symptoms, mechanisms of action, and signaling pathways involved. METHOD Research findings from experimental and clinical studies have been summarized regarding gingerols effects on the modulation of gut microbiome and its metabolites, and attenuation of disease symptoms. RESULTS Gingerols are phenolic compounds characterized by a common 3-methoxy-4-hydroxyphenyl moiety in their chemical structures, and further divided into different gingerol types, including gingerols (major), shogaols, paradols, gingerdiols, gingerdiones, and zingerones (minor). Advanced extraction techniques (e.g., ionic liquid-based-, enzyme-assisted-, microwave-assisted-, pressurized liquid-, ultrasound-assisted-, and supercritical fluid extractions) were reported as optimal alternatives to conventional methods for gingerols extraction. Research studies reported that gingerols positively modulated the composition of gut microbiome that helped to combat disease symptoms (e.g., obesity by decreasing weight gain- (Lactobacillus reuteri and Lachnospiraceae) and increasing weight loss associated-bacteria (Akkermansia, Muribaculaceae, and Alloprevotella). Gingerols intervention also ameliorated ulcerative colitis by increasing relative abundance of the beneficial bacteria (Akkermansia, Lachnospiraceae NK4A136, and Muribaculaceae_norank), and decreasing pathogenic microorganisms (Bacteroides, Parabacteroides, and Desulfovibrio). Emerging delivery systems (e.g., microcapsules, nanoparticles, nanostructured lipid carriers, nanoemulsions, and nanoliposomes) can enhance the bioavailability and therapeutic efficacy of gingerols by preserving their inherent properties and addressing challenges of stability, solubility, and absorption. CONCLUSION Gingerols are promising therapeutic agents to modulate gut microbiome (increase beneficial bacteria and inhibit pathogenic microbes), and attenuate chronic disease symptoms such as diabetes, colitis, obesity, oxidative stress, and cancer. Despite significant progress, challenges persist in transforming research findings into industrial applications, such as stability and solubility during processing and low bioavailability in the distal gut to impart desirable health benefits.
Collapse
Affiliation(s)
- Abdullah
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Naveed Ahmad
- Multan College of Food & Nutrition Sciences, Multan Medical & Dental College, Multan, Pakistan
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Wenni Tian
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Naveed Ullah Khan
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Muhammad Hussain
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hafiz Muhammad Ahsan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yahya Saud Hamed
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Rongfa Guan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
9
|
Zhou Z, Kleis L, Depetris-Chauvin A, Jaskulski S, Damerell V, Michels KB, Gigic B, Nöthlings U, Panagiotou G. Beneficial microbiome and diet interplay in early-onset colorectal cancer. EMBO Mol Med 2025; 17:9-30. [PMID: 39653811 PMCID: PMC11730345 DOI: 10.1038/s44321-024-00177-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/30/2024] [Accepted: 11/08/2024] [Indexed: 01/15/2025] Open
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed cancer and the second leading cause of cancer-related deaths worldwide. Although the risk of developing CRC increases with age, approximately 10% of newly diagnosed cases occur in individuals under the age of 50. Significant changes in dietary habits in young adults since industrialization create a favorable microenvironment for colorectal carcinogenesis. We aim here to shed light on the complex interplay between diet and gut microbiome in the pathogenesis and prevention of early-onset CRC (EO-CRC). We provide an overview of dietary risk factors associated with EO-CRC and contrast them with the general trends for CRC. We delve into gut bacteria, fungi, and phages with potential benefits against CRC and discuss the underlying molecular mechanisms. Furthermore, based on recent findings from human studies, we offer insights into how dietary modifications could potentially enhance gut microbiome composition to mitigate CRC risk. All together, we outline the current research landscape in this area and propose directions for future investigations that could pave the way for novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Zhengyuan Zhou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Jena, Germany
| | - Linda Kleis
- Institute of Nutritional and Food Sciences-Nutritional Epidemiology, University of Bonn, Friedrich-Hirzebruch-Allee 7, 53115, Bonn, Germany
| | - Ana Depetris-Chauvin
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Jena, Germany
| | - Stefanie Jaskulski
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Victoria Damerell
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Karin B Michels
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Biljana Gigic
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Ute Nöthlings
- Institute of Nutritional and Food Sciences-Nutritional Epidemiology, University of Bonn, Friedrich-Hirzebruch-Allee 7, 53115, Bonn, Germany.
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Jena, Germany.
- Friedrich Schiller University, Faculty of Biological Sciences, Jena, Germany.
- Friedrich Schiller University, Jena University Hospital, Jena, Germany.
| |
Collapse
|
10
|
Li S, Zhu S, Yu J. The role of gut microbiota and metabolites in cancer chemotherapy. J Adv Res 2024; 64:223-235. [PMID: 38013112 PMCID: PMC11464465 DOI: 10.1016/j.jare.2023.11.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND The microbiota inhabits the epithelial surfaces of hosts, which influences physiological functions from helping digest food and acquiring nutrition to regulate metabolism and shaping host immunity. With the deep insight into the microbiota, an increasing amount of research reveals that it is also involved in the initiation and progression of cancer. Intriguingly, gut microbiota can mediate the biotransformation of drugs, thereby altering their bioavailability, bioactivity, or toxicity. AIM OF REVIEW The review aims to elaborate on the role of gut microbiota and microbial metabolites in the efficacy and adverse effects of chemotherapeutics. Furthermore, we discuss the clinical potential of various ways to harness gut microbiota for cancer chemotherapy. KEY SCIENTIFIC CONCEPTS OF REVIEW Recent evidence shows that gut microbiota modulates the efficacy and toxicity of chemotherapy agents, leading to diverse host responses to chemotherapy. Thereinto, targeting the microbiota to improve efficacy and diminish the toxicity of chemotherapeutic drugs may be a promising strategy in tumor treatment.
Collapse
Affiliation(s)
- Shiyu Li
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Shuangli Zhu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
11
|
Balendra V, Rosenfeld R, Amoroso C, Castagnone C, Rossino MG, Garrone O, Ghidini M. Postbiotics as Adjuvant Therapy in Cancer Care. Nutrients 2024; 16:2400. [PMID: 39125280 PMCID: PMC11314502 DOI: 10.3390/nu16152400] [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: 06/04/2024] [Revised: 07/14/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
Postbiotics are defined as a preparation of inanimate microorganisms and/or their components that confers a health benefit to the host. They range from cell wall fragments to metabolites, bacterial lysates, extracellular vesicles, and short-chain fatty acids (SCFAs). Postbiotics may influence carcinogenesis via a variety of mechanisms. They can promote homeostatic immune responses, reduce inflammation, induce selective cytotoxicity against tumor cells, as well as the enabling the control of tumor cell proliferation and enhancing intestinal epithelial barrier function. Therefore, probiotics can serve as an adjunct strategy in anticancer treatment together with chemotherapy and immunotherapy. Up to now, the only relevant postbiotics used as interventions in oncological patients remain vitamin K molecules, with few phase-II and III trials available. In fact, postbiotics' levels are strictly dependent on the gut microbiota's composition, which may vary between individuals and can be altered under different physiological and pathological conditions. Therefore, the lack of consistent clinical evidence supporting postbiotics' efficacy is due to their poor bioavailability, short half-life, and fluctuating levels. Synbiotics, a mixture of prebiotics and probiotics, are expected to have a more homogeneous bioavailability with respect to postbiotics and may have greater potential for future development. In this review, we focus on the role of postbiotics as an adjuvant therapy in cancer treatment.
Collapse
Affiliation(s)
| | - Roberto Rosenfeld
- Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.R.); (M.G.R.); (O.G.)
| | - Chiara Amoroso
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | | | - Maria Grazia Rossino
- Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.R.); (M.G.R.); (O.G.)
| | - Ornella Garrone
- Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.R.); (M.G.R.); (O.G.)
| | - Michele Ghidini
- Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.R.); (M.G.R.); (O.G.)
| |
Collapse
|
12
|
Intarajak T, Udomchaiprasertkul W, Khoiri AN, Sutheeworapong S, Kusonmano K, Kittichotirat W, Thammarongtham C, Cheevadhanarak S. Distinct gut microbiomes in Thai patients with colorectal polyps. World J Gastroenterol 2024; 30:3336-3355. [PMID: 39086748 PMCID: PMC11287419 DOI: 10.3748/wjg.v30.i27.3336] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/30/2024] [Accepted: 05/31/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Colorectal polyps that develop via the conventional adenoma-carcinoma sequence [e.g., tubular adenoma (TA)] often progress to malignancy and are closely associated with changes in the composition of the gut microbiome. There is limited research concerning the microbial functions and gut microbiomes associated with colorectal polyps that arise through the serrated polyp pathway, such as hyperplastic polyps (HP). Exploration of microbiome alterations associated with HP and TA would improve the understanding of mechanisms by which specific microbes and their metabolic pathways contribute to colorectal carcinogenesis. AIM To investigate gut microbiome signatures, microbial associations, and microbial functions in HP and TA patients. METHODS Full-length 16S rRNA sequencing was used to characterize the gut microbiome in stool samples from control participants without polyps [control group (CT), n = 40], patients with HP (n = 52), and patients with TA (n = 60). Significant differences in gut microbiome composition and functional mechanisms were identified between the CT group and patients with HP or TA. Analytical techniques in this study included differential abundance analysis, co-occurrence network analysis, and differential pathway analysis. RESULTS Colorectal cancer (CRC)-associated bacteria, including Streptococcus gallolyticus (S. gallolyticus), Bacteroides fragilis, and Clostridium symbiosum, were identified as characteristic microbial species in TA patients. Mediterraneibacter gnavus, associated with dysbiosis and gastrointestinal diseases, was significantly differentially abundant in the HP and TA groups. Functional pathway analysis revealed that HP patients exhibited enrichment in the sulfur oxidation pathway exclusively, whereas TA patients showed dominance in pathways related to secondary metabolite biosynthesis (e.g., mevalonate); S. gallolyticus was a major contributor. Co-occurrence network and dynamic network analyses revealed co-occurrence of dysbiosis-associated bacteria in HP patients, whereas TA patients exhibited co-occurrence of CRC-associated bacteria. Furthermore, the co-occurrence of SCFA-producing bacteria was lower in TA patients than HP patients. CONCLUSION This study revealed distinct gut microbiome signatures associated with pathways of colorectal polyp development, providing insights concerning the roles of microbial species, functional pathways, and microbial interactions in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Thoranin Intarajak
- Bioinformatics Unit, Chulabhorn Royal Academy, Lak Si 10210, Bangkok, Thailand
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, and School of Information Technology, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| | | | - Ahmad Nuruddin Khoiri
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, and School of Information Technology, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| | - Sawannee Sutheeworapong
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| | - Kanthida Kusonmano
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, and School of Information Technology, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| | - Weerayuth Kittichotirat
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, and School of Information Technology, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| | - Chinae Thammarongtham
- National Center for Genetic Engineering and Biotechnology, King Mongkut's University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| | - Supapon Cheevadhanarak
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
- School of Bioresources and Technology, King Mongkut’s University of Technology Thonburi, Bank Khun Thian 10150, Bangkok, Thailand
- Fungal Biotechnology Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| |
Collapse
|
13
|
Matsui T, Morozumi T, Yamamoto Y, Kobayashi T, Takuma R, Yoneda M, Nogami A, Kessoku T, Tamura M, Nomura Y, Takahashi T, Kamata Y, Sugihara S, Arai K, Minabe M, Aoyama N, Mitsudo K, Nakajima A, Komaki M. Relationship of Metabolic Dysfunction-Associated Steatohepatitis-Related Hepatocellular Carcinoma with Oral and Intestinal Microbiota: A Cross-Sectional Pilot Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1150. [PMID: 39064580 PMCID: PMC11279156 DOI: 10.3390/medicina60071150] [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: 06/05/2024] [Revised: 07/06/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: The incidence of metabolic dysfunction-associated steatohepatitis (MASH)-related hepatocellular carcinoma (HCC) is increasing worldwide, alongside the epidemic of obesity and metabolic syndrome. Based on preliminary reports regarding the potential association of HCC and periodontitis, this study aimed to analyze the involvement of periodontal bacteria as well as the oral and intestinal bacterial flora in MASH-related HCC (MASH-HCC). Materials and Methods: Forty-one patients with MASH and nineteen with MASH-HCC participated in the study, completing survey questionnaires, undergoing periodontal examinations, and providing samples of saliva, mouth-rinsed water, feces, and peripheral blood. The oral and fecal microbiome profiles were analyzed by 16S ribosomal RNA sequencing. Bayesian network analysis was used to analyze the causation between various factors, including MASH-HCC, examinations, and bacteria. Results: The genus Fusobacterium had a significantly higher occupancy rate (p = 0.002) in the intestinal microflora of the MASH-HCC group compared to the MASH group. However, Butyricicoccus (p = 0.022) and Roseburia (p < 0.05) had significantly lower occupancy rates. The Bayesian network analysis revealed the absence of periodontal pathogenic bacteria and enteric bacteria affecting HCC. However, HCC directly affected the periodontal bacterial species Porphyromonas gingivalis, Tannerella forsythia, Fusobacterium nucleatum, and Prevotella intermedia in the saliva, as well as the genera Lactobacillus, Roseburia, Fusobacterium, Prevotella, Clostridium, Ruminococcus, Trabulsiella, and SMB53 in the intestine. Furthermore, P. gingivalis in the oral cavity directly affected the genera Lactobacillus and Streptococcus in the intestine. Conclusions: MASH-HCC directly affects periodontal pathogenic and intestinal bacteria, and P. gingivalis may affect the intestinal bacteria associated with gastrointestinal cancer.
Collapse
Affiliation(s)
- Takaaki Matsui
- Department of Periodontology, Faculty of Dentistry, Kanagawa Dental University, Yokosuka 238-8580, Japan
| | - Toshiya Morozumi
- Department of Periodontology, Faculty of Dentistry, Kanagawa Dental University, Yokosuka 238-8580, Japan
- Department of Endodontics, The Nippon Dental University School of Life Dentistry at Niigata, Niigata 951-8580, Japan
| | - Yuko Yamamoto
- Department of Dental Hygiene, Kanagawa Dental University, Junior College, Yokosuka 238-8580, Japan
| | - Takashi Kobayashi
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Ryo Takuma
- Department of Periodontology, Faculty of Dentistry, Kanagawa Dental University, Yokosuka 238-8580, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Asako Nogami
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Takaomi Kessoku
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
- Department of Palliative Medicine and Gastroenterology, International University of Health and Welfare, Narita Hospital, Narita 286-8520, Japan
| | - Muneaki Tamura
- Department of Microbiology and Immunology, Nihon University School of Dentistry, Tokyo 101-8310, Japan
| | - Yoshiaki Nomura
- Institute of Photochemistry and Photofunctional Materials, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Toru Takahashi
- Faculty of Pharmaceutical Sciences, Nihon Pharmaceutical University, Saitama 362-0806, Japan
| | - Yohei Kamata
- Department of Advanced Periodontology, Faculty of Dentistry, Kanagawa Dental University, Yokohama 221-0835, Japan
| | - Shuntaro Sugihara
- Department of Periodontology, Faculty of Dentistry, Kanagawa Dental University, Yokosuka 238-8580, Japan
| | - Kyoko Arai
- Department of Endodontics, The Nippon Dental University School of Life Dentistry at Niigata, Niigata 951-8580, Japan
| | | | - Norio Aoyama
- Department of Education Planning, Kanagawa Dental University, Yokosuka 238-8580, Japan
| | - Kenji Mitsudo
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Motohiro Komaki
- Department of Periodontology, Faculty of Dentistry, Kanagawa Dental University, Yokosuka 238-8580, Japan
| |
Collapse
|
14
|
Ozturk O, Celebi G, Duman UG, Kupcuk E, Uyanik M, Sertoglu E. Short-chain fatty acid levels in stools of patients with inflammatory bowel disease are lower than those in healthy subjects. Eur J Gastroenterol Hepatol 2024; 36:890-896. [PMID: 38829943 DOI: 10.1097/meg.0000000000002789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
OBJECTIVE Short-chain fatty acids (SCFAs) are produced when the microbiota in the large intestine cause fermentation of dietary carbohydrates and fibers. These fatty acids constitute the primary energy source of colon mucosa cells and have a protective effect in patients suffering from inflammatory bowel disease (IBD). This study aimed to compare the SCFA levels in the stools of patients with IBD and healthy controls. METHOD Healthy controls and patients with IBD aged 18 and over were included in the study. Stool samples from all patients and healthy controls were collected, and stool acetic acid, propionic acid, and butyric acid levels were measured using a gas chromatography-mass spectrometry measurement method. RESULTS In this study, 64 participants were divided into two groups: 34 were in IBD (Crohn disease and ulcerative colitis) and 30 were in healthy control group. When fecal SCFA concentrations of IBD and healthy control groups were compared, a statistically significant difference was observed between them. When the fecal SCFA concentrations of Crohn's disease and ulcerative colitis patients in the IBD group were compared, however, no statistically significant difference was observed between them. Furthermore, when the participants' diet type (carbohydrate-based, vegetable-protein-based and mixed diet) and the number of meals were compared with fecal SCFA concentrations, no statistically significant difference was observed between them. CONCLUSION In general, fecal SCFA levels in patients with IBD were lower than those in healthy controls. Moreover, diet type and the number of meals had no effect on stool SCFA levels in patients with IBD and healthy individuals.
Collapse
Affiliation(s)
| | - Gurkan Celebi
- Department of Gastroenterology, Gulhane School of Medicine, University of Health Sciences, Ankara
| | | | | | - Metin Uyanik
- Department of Biochemistry, Çorlu State Hospital, Tekirdag, Turkey
| | | |
Collapse
|
15
|
Rahman S, Lu E, Patel RK, Tsikitis VL, Martindale RG. Colorectal Disease and the Gut Microbiome: What a Surgeon Needs to Know. Surg Clin North Am 2024; 104:647-656. [PMID: 38677827 DOI: 10.1016/j.suc.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
The gut microbiome is defined as the microorganisms that reside within the gastrointestinal tract and produce a variety of metabolites that impact human health. These microbes play an intricate role in human health, and an imbalance in the gut microbiome, termed gut dysbiosis, has been implicated in the development of varying diseases. The purpose of this review is to highlight what is known about the microbiome and its impact on colorectal cancer, inflammatory bowel disease, constipation, Clostridioides difficile infection, the impact of bowel prep, and anastomotic leaks.
Collapse
Affiliation(s)
- Shahrose Rahman
- Department of Surgery, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L223, Portland, OR 97239, USA.
| | - Ethan Lu
- Department of Surgery, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L223, Portland, OR 97239, USA
| | - Ranish K Patel
- Department of Surgery, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L223, Portland, OR 97239, USA
| | - Vassiliki Liana Tsikitis
- Department of Surgery, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L223, Portland, OR 97239, USA
| | - Robert G Martindale
- Department of Surgery, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L223, Portland, OR 97239, USA
| |
Collapse
|
16
|
Masheghati F, Asgharzadeh MR, Jafari A, Masoudi N, Maleki-Kakelar H. The role of gut microbiota and probiotics in preventing, treating, and boosting the immune system in colorectal cancer. Life Sci 2024; 344:122529. [PMID: 38490297 DOI: 10.1016/j.lfs.2024.122529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/03/2023] [Accepted: 02/21/2024] [Indexed: 03/17/2024]
Abstract
The gut microbiome plays a significant role in developing colorectal cancer (CRC). The gut microbiome usually acts as a protective barrier against harmful pathogens and infections in the intestine, while also regulating inflammation by affecting the human immune system. The gut microbiota and probiotics play a role not only in intestinal inflammation associated with tumor formation but also in regulating anti-cancer immune response. As a result, they associated with tumor progression and the effectiveness of anti-cancer therapies. Research indicates that gut microbiota and probiotics can be used as biomarkers to predict the impact of immunotherapy and enhance its efficacy in treating CRC by regulating it. This review examines the importance of gut microbiota and probiotics in the development and progression of CRC, as well as their synergistic impact on anti-cancer treatments.
Collapse
Affiliation(s)
- Forough Masheghati
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Abbas Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Naser Masoudi
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of General Surgery, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Hadi Maleki-Kakelar
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
17
|
Ahmad A, Mahmood N, Raza MA, Mushtaq Z, Saeed F, Afzaal M, Hussain M, Amjad HW, Al-Awadi HM. Gut microbiota and their derivatives in the progression of colorectal cancer: Mechanisms of action, genome and epigenome contributions. Heliyon 2024; 10:e29495. [PMID: 38655310 PMCID: PMC11035079 DOI: 10.1016/j.heliyon.2024.e29495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Gut microbiota interacts with host epithelial cells and regulates many physiological functions such as genetics, epigenetics, metabolism of nutrients, and immune functions. Dietary factors may also be involved in the etiology of colorectal cancer (CRC), especially when an unhealthy diet is consumed with excess calorie intake and bad practices like smoking or consuming a great deal of alcohol. Bacteria including Fusobacterium nucleatum, Enterotoxigenic Bacteroides fragilis (ETBF), and Escherichia coli (E. coli) actively participate in the carcinogenesis of CRC. Gastrointestinal tract with chronic inflammation and immunocompromised patients are at high risk for CRC progression. Further, the gut microbiota is also involved in Geno-toxicity by producing toxins like colibactin and cytolethal distending toxin (CDT) which cause damage to double-stranded DNA. Specific microRNAs can act as either tumor suppressors or oncogenes depending on the cellular environment in which they are expressed. The current review mainly highlights the role of gut microbiota in CRC, the mechanisms of several factors in carcinogenesis, and the role of particular microbes in colorectal neoplasia.
Collapse
Affiliation(s)
- Awais Ahmad
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Nasir Mahmood
- Department of Zoology, University of Central Punjab Bahawalpur, Bahawalpur, Pakistan
| | - Muhammad Ahtisham Raza
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Zarina Mushtaq
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Farhan Saeed
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Afzaal
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muzzamal Hussain
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Hafiz Wasiqe Amjad
- International Medical School, Jinggangshan University, Ji'an, Jiangxi, China
| | | |
Collapse
|
18
|
Zheng Y, Qin C, Wen M, Zhang L, Wang W. The Effects of Food Nutrients and Bioactive Compounds on the Gut Microbiota: A Comprehensive Review. Foods 2024; 13:1345. [PMID: 38731716 PMCID: PMC11083588 DOI: 10.3390/foods13091345] [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: 03/12/2024] [Revised: 04/06/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
It is now widely recognized that gut microbiota plays a critical role not only in the development and progression of diseases, but also in its susceptibility to dietary patterns, food composition, and nutritional intake. In this comprehensive review, we have compiled the latest findings on the effects of food nutrients and bioactive compounds on the gut microbiota. The research indicates that certain components, such as unsaturated fatty acids, dietary fiber, and protein have a significant impact on the composition of bile salts and short-chain fatty acids through catabolic processes, thereby influencing the gut microbiota. Additionally, these compounds also have an effect on the ratio of Firmicutes to Bacteroides, as well as the abundance of specific species like Akkermansia muciniphila. The gut microbiota has been found to play a role in altering the absorption and metabolism of nutrients, bioactive compounds, and drugs, adding another layer of complexity to the interaction between food and gut microbiota, which often requires long-term adaptation to yield substantial outcomes. In conclusion, understanding the relationship between food compounds and gut microbiota can offer valuable insights into the potential therapeutic applications of food and dietary interventions in various diseases and health conditions.
Collapse
Affiliation(s)
- Yijun Zheng
- Clinical Pharmacy (Sino-Foreign Cooperation) Class, School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
| | - Chunyin Qin
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (C.Q.); (M.W.)
| | - Mingchun Wen
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (C.Q.); (M.W.)
| | - Liang Zhang
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (C.Q.); (M.W.)
| | - Weinan Wang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, No. 1 Xincheng Blvd, Dongguan 523808, China
| |
Collapse
|
19
|
Peng Y, Li Y, Pi Y, Yue X. Effects of almond (Armeniaca Sibirica L. Lam) polysaccharides on gut microbiota and anti-inflammatory effects on LPS-induced RAW264.7 cells. Int J Biol Macromol 2024; 263:130098. [PMID: 38342264 DOI: 10.1016/j.ijbiomac.2024.130098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/16/2024] [Accepted: 02/08/2024] [Indexed: 02/13/2024]
Abstract
The aim of this study was to investigate the prebiotic properties of the almond polysaccharide AP-1 on intestinal microorganisms by using an in vitro fecal fermentation method and its anti-inflammatory effect on lipopolysaccharide (LPS)-induced RAW264.7 cells. The results showed that during the in vitro fermentation of AP-1, the pH value of the fermentation broth decreased obviously, while the concentration of short-chain fatty acids (SCFAs) increased significantly, especially acetic acid and butyric acid. In genus level, the number of Clostridium and Megamonas increased markedly in the AP-1 group after 24 h of fermentation. After 48 h of fermentation, there was a noticeable increase in the number of beneficial genera Lactobacillaceae and Bifidobacteriaceae, and a considerable decrease in the number of pro-inflammatory genera. In addition, we found that AP-1 had no toxic effect on RAW264.7 cells. In the LPS-induced inflammation model of RAW264.7 cells, AP-1 could effectively inhibit the release of NO, regulate the level of reactive oxides (ROS), and effectively down-regulate the mRNA expression of TNF-α, IL-1β, IL-6 and iNOS. In conclusion, the almond polysaccharide AP-1 may be a functional active substance aimed at promoting intestinal health and exerting anti-inflammatory effects.
Collapse
Affiliation(s)
- Yanqi Peng
- College of Food Science, Shenyang Agricultural University, Shenyang 11086, China
| | - Yingshuo Li
- College of Food Science, Shenyang Agricultural University, Shenyang 11086, China
| | - Yuzhen Pi
- College of Food Science, Shenyang Agricultural University, Shenyang 11086, China.
| | - Xiqing Yue
- College of Food Science, Shenyang Agricultural University, Shenyang 11086, China.
| |
Collapse
|
20
|
Karim MR, Iqbal S, Mohammad S, Morshed MN, Haque MA, Mathiyalagan R, Yang DC, Kim YJ, Song JH, Yang DU. Butyrate's (a short-chain fatty acid) microbial synthesis, absorption, and preventive roles against colorectal and lung cancer. Arch Microbiol 2024; 206:137. [PMID: 38436734 DOI: 10.1007/s00203-024-03834-7] [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: 11/15/2023] [Revised: 12/28/2023] [Accepted: 01/04/2024] [Indexed: 03/05/2024]
Abstract
Butyrate, a short-chain fatty acid (SCFA) produced by bacterial fermentation of fiber in the colon, is a source of energy for colonocytes. Butyrate is essential for improving gastrointestinal (GI) health since it helps colonocyte function, reduces inflammation, preserves the gut barrier, and fosters a balanced microbiome. Human colonic butyrate producers are Gram-positive firmicutes, which are phylogenetically varied. The two most prevalent subgroups are associated with Eubacterium rectale/Roseburia spp. and Faecalibacterium prausnitzii. Now, the mechanism for the production of butyrate from microbes is a very vital topic to know. In the present study, we discuss the genes encoding the core of the butyrate synthesis pathway and also discuss the butyryl-CoA:acetate CoA-transferase, instead of butyrate kinase, which usually appears to be the enzyme that completes the process. Recently, butyrate-producing microbes have been genetically modified by researchers to increase butyrate synthesis from microbes. The activity of butyrate as a histone deacetylase inhibitor (HDACi) has led to several clinical trials to assess its effectiveness as a potential cancer treatment. Among various significant roles, butyrate is the main energy source for intestinal epithelial cells, which helps maintain colonic homeostasis. Moreover, people with non-small-cell lung cancer (NSCLC) have distinct gut microbiota from healthy adults and frequently have dysbiosis of the butyrate-producing bacteria in their guts. So, with an emphasis on colon and lung cancer, this review also discusses how the microbiome is crucial in preventing the progression of certain cancers through butyrate production. Further studies should be performed to investigate the underlying mechanisms of how these specific butyrate-producing bacteria can control both colon and lung cancer progression and prognosis.
Collapse
Affiliation(s)
- Md Rezaul Karim
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia, 7003, Bangladesh
| | - Safia Iqbal
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
- Department of Microbiology, Varendra Institute of Biosciences, Affiliated University of Rajshahi, Natore, 6400, Rajshahi, Bangladesh
| | - Shahnawaz Mohammad
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
| | - Md Niaj Morshed
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
| | - Md Anwarul Haque
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia, 7003, Bangladesh
| | - Ramya Mathiyalagan
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
| | - Deok Chun Yang
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
- Hanbangbio Inc., Yongin-Si, 17104, Gyeonggi-Do, Republic of Korea
| | - Yeon Ju Kim
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
| | - Joong Hyun Song
- Department of Veterinary International Medicine, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea.
| | - Dong Uk Yang
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea.
- AIBIOME, 6, Jeonmin-Ro 30Beon-Gil, Yuseong-Gu, Daejeon, Republic of Korea.
| |
Collapse
|
21
|
Pandey M, Bhattacharyya J. Gut microbiota and epigenetics in colorectal cancer: implications for carcinogenesis and therapeutic intervention. Epigenomics 2024; 16:403-418. [PMID: 38410915 DOI: 10.2217/epi-2023-0382] [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] [Indexed: 02/28/2024] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide. The occurrence of CRC is associated with various genetic and epigenetic mutations in intestinal epithelial cells that transform them into adenocarcinomas. There is increasing evidence indicating the gut microbiota plays a crucial role in the regulation of host physiological processes. Alterations in gut microbiota composition are responsible for initiating carcinogenesis through diverse epigenetic modifications, including histone modifications, ncRNAs and DNA methylation. This work was designed to comprehensively review recent findings to provide insight into the associations between the gut microbiota and CRC at an epigenetic level. These scientific insights can be used in the future to develop effective strategies for early detection and treatment of CRC.
Collapse
Affiliation(s)
- Monu Pandey
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi, 110016, India
- Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, 110608, India
| | - Jayanta Bhattacharyya
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi, 110016, India
- Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, 110608, India
| |
Collapse
|
22
|
Yu S, Wang X, Li Z, Jin D, Yu M, Li J, Li Y, Liu X, Zhang Q, Liu Y, Liu R, Wang X, Fang B, Zhang C, Wang R, Ren F. Solobacterium moorei promotes the progression of adenomatous polyps by causing inflammation and disrupting the intestinal barrier. J Transl Med 2024; 22:169. [PMID: 38368407 DOI: 10.1186/s12967-024-04977-3if:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/11/2024] [Indexed: 07/26/2024] Open
Abstract
BACKGROUND Adenomatous polyps (APs) with inflammation are risk factors for colorectal cancer. However, the role of inflammation-related gut microbiota in promoting the progression of APs is unknown. METHODS Sequencing of the 16S rRNA gene was conducted to identify characteristic bacteria in AP tissues and normal mucosa. Then, the roles of inflammation-related bacteria were clarified by Spearman correlation analysis. Furthermore, colorectal HT-29 cells, normal colon NCM460 cells, and azoxymethane-treated mice were used to investigate the effects of the characteristic bacteria on progression of APs. RESULTS The expression levels of inflammation-related markers (diamine oxidase, D-lactate, C-reactive protein, tumor necrosis factor-α, interleukin-6 and interleukin-1β) were increased, whereas the expression levels of anti-inflammatory factors (interleukin-4 and interleukin-10) were significantly decreased in AP patients as compared to healthy controls. Solobacterium moorei (S. moorei) was enriched in AP tissues and fecal samples, and significantly positively correlated with serum inflammation-related markers. In vitro, S. moorei preferentially attached to HT-29 cells and stimulated cell proliferation and production of pro-inflammatory factors. In vivo, the incidence of intestinal dysplasia was significantly increased in the S. moorei group. Gavage of mice with S. moorei upregulated production of pro-inflammatory factors, suppressed proliferation of CD4+ and CD8+cells, and disrupted the integrity of the intestinal barrier, thereby accelerating progression of APs. CONCLUSIONS S. moorei accelerated the progression of AP in mice via activation of the NF-κB signaling pathway, chronic low-grade inflammation, and intestinal barrier disruption. Targeted reduction of S. moorei presents a potential strategy to prevent the progression of APs.
Collapse
Affiliation(s)
- Shoujuan Yu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Xifan Wang
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, 10032, USA
| | - Ziyang Li
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Dekui Jin
- Department of General Practice, The Third Centers of Chinese PLA General Hospital, Beijing, 100039, China
| | - Mengyang Yu
- Department of General Practice, The Third Centers of Chinese PLA General Hospital, Beijing, 100039, China
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Yixuan Li
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Xiaoxue Liu
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Qi Zhang
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Yinghua Liu
- Department of Nutrition, The First Center of Chinese PLA General Hospital, Beijing, 100037, China
| | - Rong Liu
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Xiaoyu Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Bing Fang
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Chengying Zhang
- Department of General Practice, The Third Centers of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Ran Wang
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China.
| | - Fazheng Ren
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
23
|
Yu S, Wang X, Li Z, Jin D, Yu M, Li J, Li Y, Liu X, Zhang Q, Liu Y, Liu R, Wang X, Fang B, Zhang C, Wang R, Ren F. Solobacterium moorei promotes the progression of adenomatous polyps by causing inflammation and disrupting the intestinal barrier. J Transl Med 2024; 22:169. [PMID: 38368407 PMCID: PMC10874563 DOI: 10.1186/s12967-024-04977-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/11/2024] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND Adenomatous polyps (APs) with inflammation are risk factors for colorectal cancer. However, the role of inflammation-related gut microbiota in promoting the progression of APs is unknown. METHODS Sequencing of the 16S rRNA gene was conducted to identify characteristic bacteria in AP tissues and normal mucosa. Then, the roles of inflammation-related bacteria were clarified by Spearman correlation analysis. Furthermore, colorectal HT-29 cells, normal colon NCM460 cells, and azoxymethane-treated mice were used to investigate the effects of the characteristic bacteria on progression of APs. RESULTS The expression levels of inflammation-related markers (diamine oxidase, D-lactate, C-reactive protein, tumor necrosis factor-α, interleukin-6 and interleukin-1β) were increased, whereas the expression levels of anti-inflammatory factors (interleukin-4 and interleukin-10) were significantly decreased in AP patients as compared to healthy controls. Solobacterium moorei (S. moorei) was enriched in AP tissues and fecal samples, and significantly positively correlated with serum inflammation-related markers. In vitro, S. moorei preferentially attached to HT-29 cells and stimulated cell proliferation and production of pro-inflammatory factors. In vivo, the incidence of intestinal dysplasia was significantly increased in the S. moorei group. Gavage of mice with S. moorei upregulated production of pro-inflammatory factors, suppressed proliferation of CD4+ and CD8+cells, and disrupted the integrity of the intestinal barrier, thereby accelerating progression of APs. CONCLUSIONS S. moorei accelerated the progression of AP in mice via activation of the NF-κB signaling pathway, chronic low-grade inflammation, and intestinal barrier disruption. Targeted reduction of S. moorei presents a potential strategy to prevent the progression of APs.
Collapse
Affiliation(s)
- Shoujuan Yu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Xifan Wang
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, 10032, USA
| | - Ziyang Li
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Dekui Jin
- Department of General Practice, The Third Centers of Chinese PLA General Hospital, Beijing, 100039, China
| | - Mengyang Yu
- Department of General Practice, The Third Centers of Chinese PLA General Hospital, Beijing, 100039, China
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Yixuan Li
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Xiaoxue Liu
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Qi Zhang
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Yinghua Liu
- Department of Nutrition, The First Center of Chinese PLA General Hospital, Beijing, 100037, China
| | - Rong Liu
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Xiaoyu Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Bing Fang
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Chengying Zhang
- Department of General Practice, The Third Centers of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Ran Wang
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China.
| | - Fazheng Ren
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
24
|
Elahi Z, Shariati A, Bostanghadiri N, Dadgar-Zankbar L, Razavi S, Norzaee S, Vazirbani Arasi S, Darban-Sarokhalil D. Association of Lactobacillus, Firmicutes, Bifidobacterium, Clostridium, and Enterococcus with colorectal cancer in Iranian patients. Heliyon 2023; 9:e22602. [PMID: 38089982 PMCID: PMC10711133 DOI: 10.1016/j.heliyon.2023.e22602] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/08/2023] [Accepted: 11/15/2023] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the primary causes of cancer-associated deaths worldwide, and growing evidence shows that alteration in the gut microbiota may be a contributing factor to the development and progression of the disease. This study investigates the correlation between CRC and specific intestinal microbiota abundance, including Firmicutes, Lactobacillus, Enterococcus, Clostridium, and Bifidobacterium. MATERIAL AND METHODS In this study, 100 CRC samples and adjacent normal tissues were obtained from Iranian patients. Afterward, we assessed the abundance of the mentioned bacteria in matched tumor and normal tissue samples from 100 CRC patients, by TaqMan quantitative real-time polymerase chain reaction (qPCR). RESULTS Most of the patients (55 %) had grade II cancer (moderately differentiated), followed by grade III (poorly Differentiated) in 19 %, and the distribution of the tumor location was 65 % in the colon and 35 % in the rectum. Our research showed a substantial difference in the relative abundance of specific bacteria in tumors and healthy tissues. To this end, four genera of bacteria, including Bifidobacterium, Lactobacillus, Clostridium, and Firmicutes, exhibited statistically significant reductions in tumor tissues compared to adjacent normal tissue (p < 0.05). Conversely, Enterococcus demonstrated a statistically significant increase in tumor tissue samples (p < 0.05). Noteworthy, statistical analysis revealed a significant relationship between Enterococcus and prior cancer (p < 0.05). CONCLUSIONS These findings provide significant insight into the complex association between the gut microbiota and CRC and may pave the way for future research on novel screening methods, preventive measures, and therapeutic strategies targeting the gut microbiota in CRC patients.
Collapse
Affiliation(s)
- Zahra Elahi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Aref Shariati
- Student Research Committee, Khomein University of Medical Sciences, Khomein, Iran
- Molecular and Medicine Research Centre, Khomein University of Medical Sciences, Khomein, Iran
| | - Narjess Bostanghadiri
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Dadgar-Zankbar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shabnam Razavi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Norzaee
- Research Center for Environmental Health Technology, Iran University of Medical Sciences, Tehran, Iran
- Department of Environmental Health Engineering, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | | | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Mazumder K, Aktar A, Ramasamy S, Biswas B, Kerr PG, Blanchard C. Attenuating Colorectal Cancer Using Nine Cultivars of Australian Lupin Seeds: Apoptosis Induction Triggered by Mitochondrial Reactive Oxygen Species Generation and Caspases-3/7 Activation. Cells 2023; 12:2557. [PMID: 37947635 PMCID: PMC10647522 DOI: 10.3390/cells12212557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/21/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
As Australian lupin cultivars are rich sources of polyphenols, dietary fibers, high-quality proteins, and abundant bioactive compounds with significant antioxidant, antidiabetic, and anticancer activities, this research work is aimed at investigating the colon cancer alleviation activity of nine cultivars of lupin seeds on HCT116 and HT29 colon carcinoma cell lines through anti-proliferation assay, measurement of apoptosis, and identification of the mechanism of apoptosis. Nine cultivars were pre-screened for anti-proliferation of HCT116 and HT29 cells along with consideration of the impact of heat processing on cancer cell viability. Mandelup and Jurien showed significant inhibition of HCT116 cells, whereas the highest inhibition of HT29 cell proliferation was attained by Jurien and Mandelup. Processing decreased the anti-proliferation activity drastically. Lupin cultivars Mandelup, Barlock, and Jurien (dose: 300 μg/mL) induced early and late apoptosis of colon cancer cells in Annexin V-FITC assay. The mechanism of apoptosis was explored, which involves boosting of caspases-3/7 activation and intracellular reactive oxygen species (ROS) generation in HCT116 cells (Mandelup and Barlock) and HT29 cells (Jurien and Mandelup). Thus, the findings showed that lupin cultivars arrest cell cycles by inducing apoptosis of colorectal carcinoma cells triggered by elevated ROS generation and caspases-3/7 activation.
Collapse
Affiliation(s)
- Kishor Mazumder
- Department of Pharmacy, Jashore University of Science and Technology, Jashore 7408, Bangladesh
- School of Optometry and Vision Science, UNSW Medicine, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Asma Aktar
- Department of Pharmacy, Dhaka International University, Dhaka 1212, Bangladesh
| | - Sujatha Ramasamy
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Biswajit Biswas
- Department of Pharmacy, Jashore University of Science and Technology, Jashore 7408, Bangladesh
- Institute for Molecular Bioscience, Queensland University, Brisbane, QLD 4072, Australia
| | - Philip G. Kerr
- School of Biomedical Sciences and Graham Centre for Agricultural Innovation, Charles Sturt University, Boorooma St., Wagga Wagga, NSW 2650, Australia
| | - Christopher Blanchard
- School of Biomedical Sciences and Graham Centre for Agricultural Innovation, Charles Sturt University, Boorooma St., Wagga Wagga, NSW 2650, Australia
| |
Collapse
|
26
|
Bahuguna A, Dubey SK. Overview of the Mechanistic Potential of Probiotics and Prebiotics in Cancer Chemoprevention. Mol Nutr Food Res 2023; 67:e2300221. [PMID: 37552810 DOI: 10.1002/mnfr.202300221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/15/2023] [Indexed: 08/10/2023]
Abstract
Despite of strides in modern cancer therapeutic strategies, there has not been a successful cure for it until now and prognostic side effects and substantial toxicity to chemotherapy and subsequent homeostatic imbalance remains a major concern for professionals in this field. The significance of the human microbiome in the pathogenesis of cancer is being recognized, documented, and established worldwide. Probiotics and prebiotics are some of the most extensively researched approaches to modulate the microbiota for therapeutic purposes, and research on their potential to prevent and treat cancer has sparked an immense amount of interest. The characteristics of probiotics and prebiotics allow for an array of efficient applications in cancer preventive measures. Probiotics can also be administered coupled with chemotherapy and surgery to alleviate their side effects and help promote the effectiveness of chemotherapeutic drugs. Besides showing promising results they are accompanied by potential risks and controversies that may eventually result in clinical repercussions. This review emphasizes the mechanistic potential and oncosuppressive effects of probiotic and prebiotics through maintenance of intestinal barrier function, modifying innate immune system, immunomodulation, intestinal microbiota metabolism, inhibition of host cell proliferation, preventing pathogen colonization, and exerting selective cytotoxicity against tumor cells.
Collapse
Affiliation(s)
- Ananya Bahuguna
- Department of Biochemistry, C.B.S.H., G. B. Pant University of Agriculture and Technology, Pantnagar, Uttarakhand, 263145, India
| | - Shiv Kumar Dubey
- Department of Biochemistry, C.B.S.H., G. B. Pant University of Agriculture and Technology, Pantnagar, Uttarakhand, 263145, India
| |
Collapse
|
27
|
Wang X, Sun X, Chu J, Sun W, Yan S, Wang Y. Gut microbiota and microbiota-derived metabolites in colorectal cancer: enemy or friend. World J Microbiol Biotechnol 2023; 39:291. [PMID: 37653349 DOI: 10.1007/s11274-023-03742-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
Colorectal cancer (CRC) is a highly prevalent gastrointestinal cancer worldwide. Recent research has shown that the gut microbiota plays a significant role in the development of CRC. There is mounting evidence supporting the crucial contributions of bacteria-derived toxins and metabolites to cancer-related inflammation, immune imbalances, and the response to therapy. Besides, some gut microbiota and microbiota-derived metabolites have protective effects against CRC. This review aims to summarize the current studies on the effects and mechanisms of gut microbiota and microbiota-produced metabolites in the initiation, progression, and drug sensitivity/resistance of CRC. Additionally, we explore the clinical implications and future prospects of utilizing gut microbiota as innovative approaches for preventing and treating CRC.
Collapse
Affiliation(s)
- Xinyi Wang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xicai Sun
- Department of Hospital Office, Weifang People's Hospital, Weifang, China
| | - Jinjin Chu
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang, China
| | - Wenchang Sun
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang, China
| | - Shushan Yan
- Department of Gastrointestinal and Anal Diseases Surgery of the Affiliated Hospital, Weifang Medical University, Weifang, 261053, China.
| | - Yaowen Wang
- Department of Clinical Laboratory, Weifang People's Hospital, Weifang, 261041, China.
| |
Collapse
|
28
|
Shahbazi T, Bakhshi B, Sadegh Fazeli M, Derakhshan-Nezhadc E. Bacterial biomarkers: new aspects of colorectal tumors diagnosis: reality or fantasy. Eur J Cancer Prev 2023; 32:485-497. [PMID: 37477136 DOI: 10.1097/cej.0000000000000760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
As one of the most common cancers, colorectal cancer (CRC) is considered as a significant leading cause of cancer-related deaths worldwide. Gut microbiota is part of a complex microbial-based ecosystem in the human body so that changes in the microbiota could lead to a variety of diseases. A growing number of studies have shown that bacteria are both individually and collectively involved in the progression of CRC. The present review study provided a summary of some of the available data on the advantages and limitations of current CRC screening methods as well as gut biomarkers including genetic, epigenetic, and protein markers. Moreover, a summary of the applications and limitations in the detection of gut microbiota markers as well as their role in early diagnosis and timely treatment response in CRC patients was provided.
Collapse
Affiliation(s)
- Tayebe Shahbazi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University
| | - Bita Bakhshi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University
| | - Mohammad Sadegh Fazeli
- Department of Surgery, Division of Colo-Rectal Surgery, Imam Khomeini Medical Complex, Tehran University of Medical Sciences, Tehran and Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elahe Derakhshan-Nezhadc
- Department of Surgery, Division of Colo-Rectal Surgery, Imam Khomeini Medical Complex, Tehran University of Medical Sciences, Tehran and Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
29
|
Tojjari A, Choucair K, Sadeghipour A, Saeed A, Saeed A. Anti-Inflammatory and Immune Properties of Polyunsaturated Fatty Acids (PUFAs) and Their Impact on Colorectal Cancer (CRC) Prevention and Treatment. Cancers (Basel) 2023; 15:4294. [PMID: 37686570 PMCID: PMC10487099 DOI: 10.3390/cancers15174294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of death from cancer worldwide, with increasing incidence in the Western world. Diet has become the focus of research as a significant risk factor for CRC occurrence, and the role of dietary polyunsaturated fatty acids (PUFAs) has become an area of interest given their potential role in modulating inflammation, particularly in the pro-carcinogenic inflammatory environment of the colon. This work reviews the main types of PUFAs, their characteristics, structure, and physiologic role. We then highlight their potential role in preventing CRC, their signaling function vis-à-vis tumorigenic signaling, and their subsequent potential role in modulating response to different treatment modalities. We review pre-clinical and clinical data and discuss their potential use as adjunct therapies to currently existing treatment modalities. Given our understanding of PUFAs' immune and inflammation modulatory effects, we explore the possible combination of PUFAs with immune checkpoint inhibitors and other targeted therapies.
Collapse
Affiliation(s)
- Alireza Tojjari
- Division of Hematology and Oncology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA;
| | - Khalil Choucair
- Division of Hematology and Oncology, Department of Medicine, Barbara Ann Karmanos Cancer Institute, Detroit, MI 48201, USA;
| | - Arezoo Sadeghipour
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modarres University, Tehran 14115-175, Iran;
| | - Azhar Saeed
- Department of Pathology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA;
| | - Anwaar Saeed
- Division of Hematology and Oncology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA;
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
30
|
Li X, Feng J, Wang Z, Liu G, Wang F. Features of combined gut bacteria and fungi from a Chinese cohort of colorectal cancer, colorectal adenoma, and post-operative patients. Front Microbiol 2023; 14:1236583. [PMID: 37614602 PMCID: PMC10443710 DOI: 10.3389/fmicb.2023.1236583] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/20/2023] [Indexed: 08/25/2023] Open
Abstract
Colorectal cancer (CRC) accounts for the third highest morbidity burden among malignant tumors worldwide. Previous studies investigated gut microbiome changes that occur during colorectal adenomas (CRA) progression to overt CRC, thus highlighting the importance of the gut microbiome in carcinogenesis. However, few studies have examined gut microbiome characteristics across the entire spectrum, from CRC development to treatment. The study used 16S ribosomal ribonucleic acid and internal transcribed spacer amplicon sequencing to compare the composition of gut bacteria and fungi in a Chinese cohort of healthy controls (HC), CRC patients, CRA patients, and CRC postoperative patients (PP). Our analysis showed that beta diversity was significantly different among the four groups based on the gut bacterial and fungal data. A total of 51 species of bacteria and 8 species of fungi were identified in the HC, CRA, CRC, and PP groups. Correlation networks for both the gut bacteria and fungi in HC vs. CRA, HC vs. CRC, and HC vs. PP indicated some hub bacterial and fungal genera in each model, and the correlation between bacterial and fungal data indicated that a highly significant negative correlation exists among groups. Quantitative polymerase chain reaction (qPCR) analysis in a large cohort of HC, CRC, CRA, and PP patients demonstrated a significantly increasing trend of Fusobacterium nucleatum, Bifidobacterium bifidum, Candida albicans, and Saccharomyces cerevisiae in the feces of CRC patients than that of HC patients (p < 0.01). However, the abundance levels of CRA and PP were significantly lower in HC patients than those in CRC patients. Further studies are required to identify the functional consequences of the altered bacterial/fungal composition on metabolism and CRC tumorigenesis in the host.
Collapse
Affiliation(s)
- Xiaopeng Li
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Radiation Oncology, Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Jiahui Feng
- School of Life Sciences, Anhui Medical University, Hefei, China
| | - Zhanggui Wang
- Department of Radiation Oncology, Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Gang Liu
- School of Life Sciences, Anhui Medical University, Hefei, China
| | - Fan Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
31
|
Hes C, Jagoe RT. Gut microbiome and nutrition-related predictors of response to immunotherapy in cancer: making sense of the puzzle. BJC REPORTS 2023; 1:5. [PMID: 39516566 PMCID: PMC11523987 DOI: 10.1038/s44276-023-00008-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/07/2023] [Accepted: 07/05/2023] [Indexed: 11/16/2024]
Abstract
The gut microbiome is emerging as an important predictor of response to immune checkpoint inhibitor (ICI) therapy for patients with cancer. However, several nutrition-related patient characteristics, which are themselves associated with changes in gut microbiome, are also prognostic markers for ICI treatment response and survival. Thus, increased abundance of Akkermansia muciniphila, Phascolarctobacterium, Bifidobacterium and Rothia in stool are consistently associated with better response to ICI treatment. A. muciniphila is also more abundant in stool in patients with higher muscle mass, and muscle mass is a strong positive prognostic marker in cancer, including after ICI treatment. This review explores the complex inter-relations between the gut microbiome, diet and patient nutritional status and the correlations with response to ICI treatment. Different multivariate approaches, including archetypal analysis, are discussed to help identify the combinations of features which may select patients most likely to respond to ICI treatment.
Collapse
Affiliation(s)
- Cecilia Hes
- Peter Brojde Lung Cancer Centre, Segal Cancer Center, Jewish General Hospital, Montreal, QC, H3T 1E2, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, H4A 3J1, Canada
- Research Center of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, H2X 0A9, Canada
| | - R Thomas Jagoe
- Peter Brojde Lung Cancer Centre, Segal Cancer Center, Jewish General Hospital, Montreal, QC, H3T 1E2, Canada.
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
32
|
Li H, Sheng D, Jin C, Zhao G, Zhang L. Identifying and ranking causal microbial biomarkers for colorectal cancer at different cancer subsites and stages: a Mendelian randomization study. Front Oncol 2023; 13:1224705. [PMID: 37538123 PMCID: PMC10395834 DOI: 10.3389/fonc.2023.1224705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/26/2023] [Indexed: 08/05/2023] Open
Abstract
Introduction The gut microbiome is directly involved in colorectal carcinogenesis, but much of the epidemiological evidence for the effect of the gut microbiome on colorectal cancer (CRC) risk comes from observational studies, and it is unclear whether identified microbial alterations are the cause or consequence of CRC development. Methods Univariate Mendelian randomization (MR) analysis and multivariate MR analysis based on Bayesian model averaging were performed to comprehensively explore the microbial risk factors associated with CRC. The Network Module Structure Shift method was used to identify microbial biomarkers associated with CRC. Mediation analysis was used to explore the dietary habits-microbiota-CRC pathway. Results The results of the four methods showed that 9 bacteria had a robust causal relationship with the development of CRC. Among them, Streptococcus thermophilus reduced the risk of CRC; Eubacterium ventriosum and Streptococcus were beneficial bacteria of malignant tumors of colon (CC); Erysipelotrichaceae was a protective factor for malignant tumors of rectal (CR); Bacteroides ovatus was a risk factor for benign tumors. Finally, the mediation analysis revealed 10 pathways by which dietary regulation bacteria affected the risk of CRC, including alcohol consumption increased the risk of CC by reducing the abundance of Eubacterium ventriosum (mediated proportion: 43.044%), and the mediated proportion of other pathways was 7.026%-34.22%. Discussion These findings will contribute to the understanding of the different carcinogenic mechanisms of intestinal flora in the colon and rectum and the risk of tumor transformation, thereby aiding CRC prevention, early screening, and the development of future strategies to reduce CRC risk.
Collapse
Affiliation(s)
- Hongfeng Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Microbiome-X, National Institute of Health Data Science of China, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dashuang Sheng
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Microbiome-X, National Institute of Health Data Science of China, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuandi Jin
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Microbiome-X, National Institute of Health Data Science of China, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guoping Zhao
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Microbiome-X, National Institute of Health Data Science of China, Cheeloo College of Medicine, Shandong University, Jinan, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
- CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lei Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Microbiome-X, National Institute of Health Data Science of China, Cheeloo College of Medicine, Shandong University, Jinan, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
- Shandong Children’s Microbiome Center, Children’s Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
33
|
Zhou L, Jiang Z, Zhang Z, Xing J, Wang D, Tang D. Progress of gut microbiome and its metabolomics in early screening of colorectal cancer. Clin Transl Oncol 2023; 25:1949-1962. [PMID: 36790675 DOI: 10.1007/s12094-023-03097-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/18/2023] [Indexed: 02/16/2023]
Abstract
Gut microbes are widely considered to be closely associated with colorectal cancer (CRC) development. The microbiota is regarded as a potential identifier of CRC, as several studies have found great significant changes in CRC patients' microbiota and metabolic groups. Changes in microbiota, like Fusobacterium nucleatum and Bacteroides fragilis, also alter the metabolic activity of the host, promoting CRC development. In contrast, the metabolome is an intuitive discriminative biomarker as a small molecular bridge to distinguish CRC from healthy individuals due to the direct action of microbes on the host. More diagnostic microbial markers have been found, and the potential discriminatory power of microorganisms in CRC has been investigated through the combined use of biomic genomic metabolomics, bringing new ideas for screening fecal microbial markers. In this paper, we discuss the potential of microorganisms and their metabolites as biomarkers in CRC screening, hoping to provide thoughts and references for non-invasive screening of CRC.
Collapse
Affiliation(s)
- Lujia Zhou
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, 225000, People's Republic of China
| | - Zhengting Jiang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, 225000, People's Republic of China
| | - Zhilin Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, 225000, People's Republic of China
| | - Juan Xing
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, 225000, People's Republic of China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225000, People's Republic of China.
| |
Collapse
|
34
|
Zhao X, Wu H, Zhu R, Shang G, Wei J, Shang H, Tian P, Chen T, Wei H. Combination of thalidomide and Clostridium butyricum relieves chemotherapy-induced nausea and vomiting via gut microbiota and vagus nerve activity modulation. Front Immunol 2023; 14:1220165. [PMID: 37426650 PMCID: PMC10327820 DOI: 10.3389/fimmu.2023.1220165] [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: 05/10/2023] [Accepted: 05/31/2023] [Indexed: 07/11/2023] Open
Abstract
Nausea and vomiting (CINV) are distressful and widespread side effects of chemotherapy, and additional efficient regimens to alleviate CINV are urgently needed. In the present study, colorectal cancer (CRC) mice model induced by Azoxymethane (AOM)/Dextran Sodium Sulfate (DSS) was employed to evaluate the cancer suppression and CINV amelioration effect of the combination of thalidomide (THD) and Clostridium butyricum. Our results suggested that the combination of THD and C. butyricum abundantly enhanced the anticancer effect of cisplatin via activating the caspase-3 apoptosis pathway, and also ameliorated CINV via inhibiting the neurotransmitter (e.g., 5-HT and tachykinin 1) and its receptor (e.g., 5-HT3R and NK-1R) in brain and colon. Additionally, the combination of THD and C. butyricum reversed the gut dysbacteriosis in CRC mice by increasing the abundance of Clostridium, Lactobacillus, Bifidobacterium, and Ruminococcus at the genus level, and also led to increased expression of occludin and Trek1 in the colon, while decreased expression of TLR4, MyD88, NF-κB, and HDAC1, as well as the mRNA level of IL-6, IL-1β, and TNF-α. In all, these results suggest that the combination of THD and C. butyricum had good efficacy in enhancing cancer treatments and ameliorating CINV, which thus provides a more effective strategy for the treatment of CRC.
Collapse
Affiliation(s)
- Xuanqi Zhao
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Heng Wu
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Ruizhe Zhu
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| | | | - Jing Wei
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Haitao Shang
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Puyuan Tian
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tingtao Chen
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Hong Wei
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
35
|
Heng D, Zhang M, Yuan Y, Qiu X. Alteration of Colonic Bacterial and Fungal Composition and Their Inter- and Intra-Kingdom Interaction in Patients with Adenomas with Low-Grade Dysplasia. Microorganisms 2023; 11:1327. [PMID: 37317301 PMCID: PMC10223777 DOI: 10.3390/microorganisms11051327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 06/16/2023] Open
Abstract
Colorectal cancer (CRC) develops from pre-cancerous cellular lesions in the gut epithelium and mainly originates from specific types of colonic adenomas with dysplasia. However, gut microbiota signatures among sampling sites in patients with colorectal adenomas with low-grade dysplasia (ALGD) and normal control (NC) remain uncharacterized. To characterize gut microbial and fungal profiles in ALGD and normal colorectal mucosa tissues. We used 16S and ITS1-2 rRNA gene sequencing and bioinformatics analysis on the microbiota of ALGD and normal colorectal mucosa from 40 subjects. Bacterial sequences in the ALGD group showed an increase in Rhodobacterales, Thermales, Thermaceae, Rhodobacteraceae, and several genera, including Thermus, Paracoccus, Sphingobium, and Pseudomonas, compared to the NC group. Fungal sequences in the ALGD group showed an increase in Helotiales, Leotiomycetes, and Basidiomycota, while several orders, families, and genera, including Verrucariales, Russulales, and Trichosporonales, were decreased. The study found various interactions between intestinal bacteria and fungi. The bacterial functional analysis showed increased glycogen and vanillin degradation pathways in the ALGD group. Meanwhile, the fungal functional analysis showed a decrease in pathways related to the biosynthesis of gondoate and stearate, as well as degradation of glucose, starch, glycogen, sucrose, L-tryptophan, and pantothenate, and an increase in the octane oxidation pathway in the ALGD group. The mucosal microbiota in ALGD exhibits altered fungal and microbial composition compared to the NC mucosa, potentially contributing to the development of intestinal cancer by regulating specific metabolic pathways. Therefore, these changes in microbiota and metabolic pathways may be potential markers for diagnosing and treating colorectal adenoma and carcinoma.
Collapse
Affiliation(s)
- Ding Heng
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing 210029, China; (D.H.); (M.Z.)
| | - Min Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing 210029, China; (D.H.); (M.Z.)
| | - Yuhan Yuan
- Department of Endoscopic Center, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing 210029, China;
| | - Xinyun Qiu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing 210029, China; (D.H.); (M.Z.)
- F. Widjaja Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
36
|
Gu M, Yin W, Zhang J, Yin J, Tang X, Ling J, Tang Z, Yin W, Wang X, Ni Q, Zhu Y, Chen T. Role of gut microbiota and bacterial metabolites in mucins of colorectal cancer. Front Cell Infect Microbiol 2023; 13:1119992. [PMID: 37265504 PMCID: PMC10229905 DOI: 10.3389/fcimb.2023.1119992] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/03/2023] [Indexed: 06/03/2023] Open
Abstract
Colorectal cancer (CRC) is a major health burden, accounting for approximately 10% of all new cancer cases worldwide. Accumulating evidence suggests that the crosstalk between the host mucins and gut microbiota is associated with the occurrence and development of CRC. Mucins secreted by goblet cells not only protect the intestinal epithelium from microorganisms and invading pathogens but also provide a habitat for commensal bacteria. Conversely, gut dysbiosis results in the dysfunction of mucins, allowing other commensals and their metabolites to pass through the intestinal epithelium, potentially triggering host responses and the subsequent progression of CRC. In this review, we summarize how gut microbiota and bacterial metabolites regulate the function and expression of mucin in CRC and novel treatment strategies for CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiangjun Wang
- Department of General Surgery, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Qing Ni
- Department of General Surgery, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Yunxiang Zhu
- Department of General Surgery, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Tuo Chen
- Department of General Surgery, Affiliated Hospital of Yangzhou University, Yangzhou, China
| |
Collapse
|
37
|
Pu W, Zhang H, Zhang T, Guo X, Wang X, Tang S. Inhibitory effects of Clostridium butyricum culture and supernatant on inflammatory colorectal cancer in mice. Front Immunol 2023; 14:1004756. [PMID: 37081884 PMCID: PMC10111964 DOI: 10.3389/fimmu.2023.1004756] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 03/16/2023] [Indexed: 04/07/2023] Open
Abstract
Clostridium butyricum (CB) is a spore-forming, gram-positive and obligate anaerobic rod bacterium. CB can modulate the composition of the gut microbiome and promote the growth of beneficial microbes in the intestine by generating short-chain fatty acids (SCFAs), which in turn protect against colitis and prevents the formation of inflammatory-associated colorectal cancer (CRC) by ameliorating colon inflammatory processes. Yet, it remains unclear whether the culture and supernatant of CB could directly influence inflammatory CRC in mice. In this study, azoxymethane (AOM)+dextran sodium sulphate (DSS) was used to induce CRC model in C57BL/6 mice. Next, the serum levels of inflammatory cytokines, including interleukin-6 (IL-6), interleukin-10 (IL-10), and cytokines TNF-α, were measured and the pathohistological examination of the large intestine was performed. Both CB culture and supernatant were found to have anti-inflammatory properties. Subsequently, Western blot and Real-Time Quantitative PCR (RT-qPCR) revealed that CB and supernatant regulate the NF-κB/p65 pathway to inhibit the development and progression of inflammatory CRC in AOM+DSS-treated mice, which could be due to the high levels of butyric acid in the supernatant.
Collapse
Affiliation(s)
- Wenfeng Pu
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
- Department of Gastroenterology, Nan Chong Central Hospital, the Second Affiliated Hospital of North Sichuan Medical College, Sichuan, Nanchong, China
| | - Hong Zhang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Sichuan, Nanchong, China
- Department of Gastroenterology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Zhang
- Department of Gastroenterology, Nan Chong Central Hospital, the Second Affiliated Hospital of North Sichuan Medical College, Sichuan, Nanchong, China
| | - Xiaoguang Guo
- Department of Pathology, Nan Chong Central Hospital, the Second Affiliated Hospital of North Sichuan Medical College, Sichuan, Nanchong, China
| | - Xiaoqing Wang
- Department of Nucler Medicine, Nan Chong Central Hospital, the Second Affiliated Hospital of North Sichuan Medical College, Sichuan, Nanchong, China
| | - Shaohui Tang
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
- *Correspondence: Shaohui Tang,
| |
Collapse
|
38
|
Singh S, Sharma P, Sarma DK, Kumawat M, Tiwari R, Verma V, Nagpal R, Kumar M. Implication of Obesity and Gut Microbiome Dysbiosis in the Etiology of Colorectal Cancer. Cancers (Basel) 2023; 15:1913. [PMID: 36980799 PMCID: PMC10047102 DOI: 10.3390/cancers15061913] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/12/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
The complexity and variety of gut microbiomes within and among individuals have been extensively studied in recent years in connection to human health and diseases. Our growing understanding of the bidirectional communication between metabolic diseases and the gut microbiome has also highlighted the significance of gut microbiome dysbiosis in the genesis and development of obesity-related cancers. Therefore, it is crucial to comprehend the possible role of the gut microbiota in the crosstalk between obesity and colorectal cancer (CRC). Through the induction of gut microbial dysbiosis, gut epithelial barrier impairment, metabolomic dysregulation, chronic inflammation, or dysregulation in energy harvesting, obesity may promote the development of colorectal tumors. It is well known that strategies for cancer prevention and treatment are most effective when combined with a healthy diet, physical activity, and active lifestyle choices. Recent studies also suggest that an improved understanding of the complex linkages between the gut microbiome and various cancers as well as metabolic diseases can potentially improve cancer treatments and overall outcomes. In this context, we herein review and summarize the clinical and experimental evidence supporting the functional role of the gut microbiome in the pathogenesis and progression of CRC concerning obesity and its metabolic correlates, which may pave the way for the development of novel prognostic tools for CRC prevention. Therapeutic approaches for restoring the microbiome homeostasis in conjunction with cancer treatments are also discussed herein.
Collapse
Affiliation(s)
- Samradhi Singh
- Indian Council of Medical Research-National Institute for Research in Environmental Health, Bhopal 462030, India
| | - Poonam Sharma
- Indian Council of Medical Research-National Institute for Research in Environmental Health, Bhopal 462030, India
| | - Devojit Kumar Sarma
- Indian Council of Medical Research-National Institute for Research in Environmental Health, Bhopal 462030, India
| | - Manoj Kumawat
- Indian Council of Medical Research-National Institute for Research in Environmental Health, Bhopal 462030, India
| | - Rajnarayan Tiwari
- Indian Council of Medical Research-National Institute for Research in Environmental Health, Bhopal 462030, India
| | - Vinod Verma
- Stem Cell Research Centre, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow 226014, India
| | - Ravinder Nagpal
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32302, USA
| | - Manoj Kumar
- Indian Council of Medical Research-National Institute for Research in Environmental Health, Bhopal 462030, India
| |
Collapse
|
39
|
Niekamp P, Kim CH. Microbial Metabolite Dysbiosis and Colorectal Cancer. Gut Liver 2023; 17:190-203. [PMID: 36632785 PMCID: PMC10018301 DOI: 10.5009/gnl220260] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/09/2022] [Accepted: 08/18/2022] [Indexed: 01/13/2023] Open
Abstract
The global burden of colorectal cancer (CRC) is expected to continuously increase. Through research performed in the past decades, the effects of various environmental factors on CRC development have been well identified. Diet, the gut microbiota and their metabolites are key environmental factors that profoundly affect CRC development. Major microbial metabolites with a relevance for CRC prevention and pathogenesis include dietary fiber-derived short-chain fatty acids, bile acid derivatives, indole metabolites, polyamines, trimethylamine-N-oxide, formate, and hydrogen sulfide. These metabolites regulate various cell types in the intestine, leading to an altered intestinal barrier, immunity, chronic inflammation, and tumorigenesis. The physical, chemical, and metabolic properties of these metabolites along with their distinct functions to trigger host receptors appear to largely determine their effects in regulating CRC development. In this review, we will discuss the current advances in our understanding of the major CRC-regulating microbial metabolites, focusing on their production and interactive effects on immune responses and tumorigenesis in the colon.
Collapse
Affiliation(s)
- Patrick Niekamp
- Department of Pathology and Mary H. Weiser Food Allergy Center, Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Chang H. Kim
- Department of Pathology and Mary H. Weiser Food Allergy Center, Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI, USA
| |
Collapse
|
40
|
Valciukiene J, Strupas K, Poskus T. Tissue vs. Fecal-Derived Bacterial Dysbiosis in Precancerous Colorectal Lesions: A Systematic Review. Cancers (Basel) 2023; 15:1602. [PMID: 36900392 PMCID: PMC10000868 DOI: 10.3390/cancers15051602] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/19/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Alterations in gut microbiota play a pivotal role in the adenoma-carcinoma sequence. However, there is still a notable lack of the correct implementation of tissue and fecal sampling in the setting of human gut microbiota examination. This study aimed to review the literature and to consolidate the current evidence on the use of mucosa and a stool-based matrix investigating human gut microbiota changes in precancerous colorectal lesions. A systematic review of papers from 2012 until November 2022 published on the PubMed and Web of Science databases was conducted. The majority of the included studies have significantly associated gut microbial dysbiosis with premalignant polyps in the colorectum. Although methodological differences hampered the precise fecal and tissue-derived dysbiosis comparison, the analysis revealed several common characteristics in stool-based and fecal-derived gut microbiota structures in patients with colorectal polyps: simple or advanced adenomas, serrated lesions, and carcinomas in situ. The mucosal samples considered were more relevant for the evaluation of microbiota's pathophysiological involvement in CR carcinogenesis, while non-invasive stool sampling could be beneficial for early CRC detection strategies in the future. Further studies are required to identify and validate mucosa-associated and luminal colorectal microbial patterns and their role in CRC carcinogenesis, as well as in the clinical setting of human microbiota studies.
Collapse
Affiliation(s)
- Jurate Valciukiene
- Clinic of Gastroenterology, Nephro-Urology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | | | | |
Collapse
|
41
|
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related death in the United States. Once limited to older populations, the incidence of CRC in patients under the age of 50 years is increasing and the etiology for this is uncertain. One hypothesis lies on the impact of the intestinal microbiome. The intestinal microbiome, composed primarily of bacteria but also viruses, fungi, and archaea, has been shown to regulate CRC development and progression both in vitro and in vivo. In this review, the role and intersection of the bacterial microbiome in various stages of clinical CRC development and management are discussed beginning with CRC screening. Various mechanisms whereby the microbiome has been shown to modulate CRC development including the influence of diet on the microbiome, bacterial-induced injury to the colonic epithelium, bacterial-produced toxins, and alteration of normal cancer immunosurveillance by the microbiome are discussed. Finally, the influence of microbiome on the response of CRC to treatment is discussed while highlighting ongoing clinical trials. The complexities of the microbiome and its role in CRC development and progression have become apparent and will require ongoing commitment to translate laboratory findings into meaningful clinical results that will aid more than 150,000 patients that develop CRC every year.
Collapse
Affiliation(s)
- Ryan M. Thomas
- Department of Surgery, University of Florida, Gainesville, Florida
| |
Collapse
|
42
|
Bioaccessibility and Bioavailability of Diet Polyphenols and Their Modulation of Gut Microbiota. Int J Mol Sci 2023; 24:ijms24043813. [PMID: 36835225 PMCID: PMC9961503 DOI: 10.3390/ijms24043813] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
It is generally accepted that diet-derived polyphenols are bioactive compounds with several potentially beneficial effects on human health. In general, polyphenols have several chemical structures, and the most representative are flavonoids, phenolic acids, and stilbenes. It should be noted that the beneficial effects of polyphenols are closely related to their bioavailability and bioaccessibility, as many of them are rapidly metabolized after administration. Polyphenols-with a protective effect on the gastrointestinal tract-promote the maintenance of the eubiosis of the intestinal microbiota with protective effects against gastric and colon cancers. Thus, the benefits obtained from dietary supplementation of polyphenols would seem to be mediated by the gut microbiota. Taken at certain concentrations, polyphenols have been shown to positively modulate the bacterial component, increasing Lactiplantibacillus spp. and Bifidobacterium spp. involved in the protection of the intestinal barrier and decreasing Clostridium and Fusobacterium, which are negatively associated with human well-being. Based on the diet-microbiota-health axis, this review aims to describe the latest knowledge on the action of dietary polyphenols on human health through the activity of the gut microbiota and discusses micro-encapsulation of polyphenols as a strategy to improve the microbiota.
Collapse
|
43
|
Lu Y, Zhao YC, Chang-Claude J, Gruber SB, Gsur A, Offit K, Vodickova L, Woods MO, Nguyen LH, Wade KH, Carreras-Torres R, Moreno V, Buchanan DD, Cotterchio M, Chan AT, Phipps AI, Peters U, Song M. Genetic Predictors for Fecal Propionate and Butyrate-Producing Microbiome Pathway Are Not Associated with Colorectal Cancer Risk: A Mendelian Randomization Analysis. Cancer Epidemiol Biomarkers Prev 2023; 32:281-286. [PMID: 36512731 PMCID: PMC9905300 DOI: 10.1158/1055-9965.epi-22-0861] [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: 08/08/2022] [Revised: 10/14/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Mechanistic data indicate the benefit of short-chain fatty acids (SCFA) produced by gut microbial fermentation of fiber on colorectal cancer, but direct epidemiologic evidence is limited. A recent study identified SNPs for two SCFA traits (fecal propionate and butyrate-producing microbiome pathway PWY-5022) in Europeans and showed metabolic benefits. METHODS We conducted a two-sample Mendelian randomization analysis of the genetic instruments for the two SCFA traits (three SNPs for fecal propionate and nine for PWY-5022) in relation to colorectal cancer risk in three large European genetic consortia of 58,131 colorectal cancer cases and 67,347 controls. We estimated the risk of overall colorectal cancer and conducted subgroup analyses by sex, age, and anatomic subsites of colorectal cancer. RESULTS We did not observe strong evidence for an association of the genetic predictors for fecal propionate levels and the abundance of PWY-5022 with the risk of overall colorectal cancer, colorectal cancer by sex, or early-onset colorectal cancer (diagnosed at <50 years), with no evidence of heterogeneity or pleiotropy. When assessed by tumor subsites, we found weak evidence for an association between PWY-5022 and risk of rectal cancer (OR per 1-SD, 0.95; 95% confidence intervals, 0.91-0.99; P = 0.03) but it did not surpass multiple testing of subgroup analysis. CONCLUSIONS Genetic instruments for fecal propionate levels and the abundance of PWY-5022 were not associated with colorectal cancer risk. IMPACT Fecal propionate and PWY-5022 may not have a substantial influence on colorectal cancer risk. Future research is warranted to comprehensively investigate the effects of SCFA-producing bacteria and SCFAs on colorectal cancer risk.
Collapse
Affiliation(s)
- Yujia Lu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Yu Chen Zhao
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- University Cancer Center Hamburg (UCCH), Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephen B. Gruber
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Andrea Gsur
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Kenneth Offit
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Ludmila Vodickova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Michael O. Woods
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John’s, Canada
| | - Long H. Nguyen
- Division of Gastroenterology, Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kaitlin H. Wade
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Robert Carreras-Torres
- Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908, Barcelona, Spain
- Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), Salt, 17190, Girona, Spain
| | - Victor Moreno
- Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908, Barcelona, Spain
- Oncology Data Analytics Program, Catalan Institute of Oncology, L’Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Daniel D. Buchanan
- Colorectal Oncogenomic Group, Department of Clinical Pathology, University of Melbourne, Parkville, Victoria, Australia
- Victorian Comprehensive Cancer Centre, University of Melbourne Centre for Cancer Research, Parkville, Victoria, Australia
- Genetic Medicine and Family Clinic, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Michelle Cotterchio
- Prevention and Cancer Control, Clinical Institutes and Quality Programs, Ontario Health (Cancer Care Ontario), Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Andrew T. Chan
- Division of Gastroenterology, Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
| | - Amanda I. Phipps
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA 98195, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Ulrike Peters
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA 98195, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Mingyang Song
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Division of Gastroenterology, Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
44
|
Zou S, Qin B, Yang Z, Wang W, Zhang J, Zhang Y, Meng M, Feng J, Xie Y, Fang L, Xiao L, Zhang P, Meng X, Choi HH, Wen W, Pan Q, Ghesquière B, Lan P, Lee MH, Fang L. CSN6 Mediates Nucleotide Metabolism to Promote Tumor Development and Chemoresistance in Colorectal Cancer. Cancer Res 2023; 83:414-427. [PMID: 36512632 DOI: 10.1158/0008-5472.can-22-2145] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 10/10/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Metabolic reprogramming can contribute to colorectal cancer progression and therapy resistance. Identification of key regulators of colorectal cancer metabolism could provide new approaches to improve treatment and reduce recurrence. Here, we demonstrate a critical role for the COP9 signalosome subunit CSN6 in rewiring nucleotide metabolism in colorectal cancer. Transcriptomic analysis of colorectal cancer patient samples revealed a correlation between CSN6 expression and purine and pyrimidine metabolism. A colitis-associated colorectal cancer model established that Csn6 intestinal conditional deletion decreased tumor development and altered nucleotide metabolism. CSN6 knockdown increased the chemosensitivity of colorectal cancer cells in vitro and in vivo, which could be partially reversed with nucleoside supplementation. Isotope metabolite tracing showed that CSN6 loss reduced de novo nucleotide synthesis. Mechanistically, CSN6 upregulated purine and pyrimidine biosynthesis by increasing expression of PHGDH, a key enzyme in the serine synthesis pathway. CSN6 inhibited β-Trcp-mediated DDX5 polyubiquitination and degradation, which in turn promoted DDX5-mediated PHGDH mRNA stabilization, leading to metabolic reprogramming and colorectal cancer progression. Butyrate treatment decreased CSN6 expression and improved chemotherapy efficacy. These findings unravel the oncogenic role of CSN6 in regulating nucleotide metabolism and chemosensitivity in colorectal cancer. SIGNIFICANCE CSN6 deficiency inhibits colorectal cancer development and chemoresistance by downregulating PHGDH to block nucleotide biosynthesis, providing potential therapeutic targets to improve colorectal cancer treatment.
Collapse
Affiliation(s)
- Shaomin Zou
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Baifu Qin
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Ziqing Yang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Wencong Wang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Jieping Zhang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Yijing Zhang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Manqi Meng
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Junyan Feng
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Yunling Xie
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Ling Fang
- Instrumental Analysis and Research Center, Sun Yat-sen University, Guangzhou, China
| | - Lishi Xiao
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Peng Zhang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Xiangqi Meng
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Hyun Ho Choi
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Weijie Wen
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Qihao Pan
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Bart Ghesquière
- Metabolomics Core Facility, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Ping Lan
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China.,Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mong-Hong Lee
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Lekun Fang
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangzhou, China.,Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
45
|
Pandey H, Tang DWT, Wong SH, Lal D. Gut Microbiota in Colorectal Cancer: Biological Role and Therapeutic Opportunities. Cancers (Basel) 2023; 15:cancers15030866. [PMID: 36765824 PMCID: PMC9913759 DOI: 10.3390/cancers15030866] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Colorectal cancer (CRC) is the second-leading cause of cancer-related deaths worldwide. While CRC is thought to be an interplay between genetic and environmental factors, several lines of evidence suggest the involvement of gut microbiota in promoting inflammation and tumor progression. Gut microbiota refer to the ~40 trillion microorganisms that inhabit the human gut. Advances in next-generation sequencing technologies and metagenomics have provided new insights into the gut microbial ecology and have helped in linking gut microbiota to CRC. Many studies carried out in humans and animal models have emphasized the role of certain gut bacteria, such as Fusobacterium nucleatum, enterotoxigenic Bacteroides fragilis, and colibactin-producing Escherichia coli, in the onset and progression of CRC. Metagenomic studies have opened up new avenues for the application of gut microbiota in the diagnosis, prevention, and treatment of CRC. This review article summarizes the role of gut microbiota in CRC development and its use as a biomarker to predict the disease and its potential therapeutic applications.
Collapse
Affiliation(s)
- Himani Pandey
- Redcliffe Labs, Electronic City, Noida 201301, India
| | - Daryl W. T. Tang
- School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore
| | - Sunny H. Wong
- Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Correspondence: (S.H.W.); (D.L.)
| | - Devi Lal
- Department of Zoology, Ramjas College, University of Delhi, Delhi 110007, India
- Correspondence: (S.H.W.); (D.L.)
| |
Collapse
|
46
|
Masood M, Nasser MI. Gut microbial metabolites and colorectal cancer. MICROBIAL BIOMOLECULES 2023:353-373. [DOI: 10.1016/b978-0-323-99476-7.00011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
47
|
Nami Y, Hejazi S, Geranmayeh MH, Shahgolzari M, Yari Khosroushahi A. Probiotic immunonutrition impacts on colon cancer immunotherapy and prevention. Eur J Cancer Prev 2023; 32:30-47. [PMID: 36134612 DOI: 10.1097/cej.0000000000000738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The important role of the immune system in treating cancer has attracted the attention of researchers to the emergence of oncology research. Immunotherapy has shown that the immune system is important in the fight against cancer. The challenge has led researchers to analyze the impact of immunotherapy on improving the status of the immune system, modifying the resulting safety response, reducing toxicity, and improving the results. This study aimed to discuss the potential mechanisms of probiotics in preventing colon cancer. The mechanisms include the change in intestinal microbiota, the metabolic activity of microbiota, the binding and degradation of the carcinogenic compounds present in the lumen of the intestine, the production of compounds with anticancer activity, immune system modification, intestinal dysfunction, changes in host physiology, and inhibition of cell proliferation and induction of apoptosis in cancerous cells. By contrast, very few reports have shown the harmful effects of oral probiotic supplements. According to available evidence, further studies on probiotics are needed, especially in identifying bacterial species with anticancer potential, studying the survival of the strains after passing the digestive tract, reviewing potential side effects in people with a weak immune system, and ultimately consuming and repeating its use. This study emphasizes that the nutritional formula can modulate inflammatory and immune responses in cancer patients. This effect reduces acute toxicity, although the pathways and measurement of this immune response are unclear. Nutrition safety is an emerging field in oncology, and further research is required.
Collapse
Affiliation(s)
- Yousef Nami
- Department of Food Biotechnology, Branch for Northwest & West Region, Agricultural Biotechnology Research Institute of Iran, Agricultural Research, Education and Extension Organization (AREEO)
| | - Salva Hejazi
- Department of Medicine, Student Research Committee, Tabriz University of Medical Sciences
| | - Mohammad Hossein Geranmayeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences
| | - Mehdi Shahgolzari
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences
- Biotechnology Research Center, Tabriz University of Medical Sciences
| | - Ahmad Yari Khosroushahi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
48
|
Systematic Review: Contribution of the Gut Microbiome to the Volatile Metabolic Fingerprint of Colorectal Neoplasia. Metabolites 2022; 13:metabo13010055. [PMID: 36676980 PMCID: PMC9865897 DOI: 10.3390/metabo13010055] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Colorectal cancer (CRC) has been associated with changes in volatile metabolic profiles in several human biological matrices. This enables its non-invasive detection, but the origin of these volatile organic compounds (VOCs) and their relation to the gut microbiome are not yet fully understood. This systematic review provides an overview of the current understanding of this topic. A systematic search using PubMed, Embase, Medline, Cochrane Library, and the Web of Science according to PRISMA guidelines resulted in seventy-one included studies. In addition, a systematic search was conducted that identified five systematic reviews from which CRC-associated gut microbiota data were extracted. The included studies analyzed VOCs in feces, urine, breath, blood, tissue, and saliva. Eight studies performed microbiota analysis in addition to VOC analysis. The most frequently reported dysregulations over all matrices included short-chain fatty acids, amino acids, proteolytic fermentation products, and products related to the tricarboxylic acid cycle and Warburg metabolism. Many of these dysregulations could be related to the shifts in CRC-associated microbiota, and thus the gut microbiota presumably contributes to the metabolic fingerprint of VOC in CRC. Future research involving VOCs analysis should include simultaneous gut microbiota analysis.
Collapse
|
49
|
Shuwen H, Yinhang W, Xingming Z, Jing Z, Jinxin L, Wei W, Kefeng D. Using whole-genome sequencing (WGS) to plot colorectal cancer-related gut microbiota in a population with varied geography. Gut Pathog 2022; 14:50. [PMID: 36578080 PMCID: PMC9795735 DOI: 10.1186/s13099-022-00524-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a multifactorial disease with genetic and environmental factors. Regional differences in risk factors are an important reason for the different incidences of CRC in different regions. OBJECTIVE The goal was to clarify the intestinal microbial composition and structure of CRC patients in different regions and construct CRC risk prediction models based on regional differences. METHODS A metagenomic dataset of 601 samples from 6 countries in the GMrepo and NCBI databases was collected. All whole-genome sequencing (WGS) data were annotated for species by MetaPhlAn2. We obtained the relative abundance of species composition at the species level and genus level. The MicrobiotaProcess package was used to visualize species composition and PCA. LEfSe analysis was used to analyze the differences in the datasets in each region. Spearman correlation analysis was performed for CRC differential species. Finally, the CRC risk prediction model was constructed and verified in each regional dataset. RESULTS The composition of the intestinal bacterial community varied in different regions. Differential intestinal bacteria of CRC in different regions are inconsistent. There was a common diversity of bacteria in all six countries, such as Peptostreptococcus stomatis and Fusobacterium nucleatum at the species level. Peptostreptococcus stomatis (species level) and Peptostreptococcus (genus level) are important CRC-related bacteria that are related to other bacteria in different regions. Region has little influence on the accuracy of the CRC risk prediction model. Peptostreptococcus stomatis is an important variable in CRC risk prediction models in all regions. CONCLUSION Peptostreptococcus stomatis is a common high-risk pathogen of CRC worldwide, and it is an important variable in CRC risk prediction models in all regions. However, regional differences in intestinal bacteria had no significant impact on the accuracy of the CRC risk prediction model.
Collapse
Affiliation(s)
- Han Shuwen
- grid.412465.0Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Building 6 Room 2018, Hangzhou, 310009 Zhejiang China ,grid.413679.e0000 0004 0517 0981Huzhou Central Hospital, Huzhou, Zhejiang China
| | - Wu Yinhang
- grid.413679.e0000 0004 0517 0981Huzhou Central Hospital, Huzhou, Zhejiang China
| | - Zhao Xingming
- grid.8547.e0000 0001 0125 2443Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Zhuang Jing
- grid.413679.e0000 0004 0517 0981Huzhou Central Hospital, Huzhou, Zhejiang China
| | - Liu Jinxin
- grid.8547.e0000 0001 0125 2443Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Wu Wei
- grid.413679.e0000 0004 0517 0981Huzhou Central Hospital, Huzhou, Zhejiang China
| | - Ding Kefeng
- grid.412465.0Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Building 6 Room 2018, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center Zhejiang University, Hangzhou, Zhejiang China
| |
Collapse
|
50
|
Abstract
The gut microbiota (GM) is associated with colorectal cancer (CRC) development. However, studies demonstrating the role of GM in CRC are limited to metagenomic analyses. These studies lack direct evidence proving that the candidate strains are involved in CRC, and isolated probiotics for bacteriotherapy. Therefore, to identify novel GM with anti-CRC activity, we previously isolated gut bacteria from the feces of healthy individuals, screened the isolated GM's anti-CRC activity, and discovered that cell-free supernatants of GM isolates demonstrated antiproliferative activity against CRC cells. Here, our study identified one of them as Eubacterium callanderi and chose it for further study because the genus Eubacterium has been suggested to contribute to various aspects of gut health; however, the functions are unknown. First, we confirmed that E. callanderi cell-free supernatant (EcCFS) exerted antiproliferative activity-by inducing apoptosis and cell cycle arrest-that was dose-dependent and specific to cancer cell lines. Next, we discovered that EcCFS active molecules were heat stable and protease insensitive. High-performance liquid chromatography analysis revealed that EcCFS contained high butyrate concentrations possessing anticancer activity. Additionally, gas chromatography-mass spectrometry analysis of the aqueous phase of ethyl acetate-extracted EcCFS and an antiproliferation assay of the aqueous phase and 4-aminobutanoic acid (GABA) suggested that GABA is a possible anti-CRC agent. Finally, in the CT26 allograft mouse model, E. callanderi oral administration and EcCFS peri-tumoral injection inhibited tumor growth in vivo. Therefore, our study reveals that E. callanderi has an anti-CRC effect and suggests that it may be a potential candidate for developing probiotics to control CRC. IMPORTANCE The gut microbiota has been reported to be involved in colorectal cancer, as suggested by metagenomic analysis. However, metagenomic analysis has limitations, such as bias in the analysis and the absence of bacterial resources for follow-up studies. Therefore, we attempted to discover gut microorganisms that are related to colorectal cancer using the culturomics method. In this study, we discovered that Eubacterium callanderi possesses anti-colorectal cancer activity in vitro and in vivo, suggesting that E. callanderi could be used in bacteriotherapy for colorectal cancer treatment.
Collapse
|