51
|
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
|
52
|
Sawhney SS, Thänert R, Thänert A, Hall-Moore C, Ndao IM, Mahmud B, Warner BB, Tarr PI, Dantas G. Gut microbiome evolution from infancy to 8 years of age. Nat Med 2025:10.1038/s41591-025-03610-0. [PMID: 40175737 DOI: 10.1038/s41591-025-03610-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 02/24/2025] [Indexed: 04/04/2025]
Abstract
The human gut microbiome is most dynamic in early life. Although sweeping changes in taxonomic architecture are well described, it remains unknown how, and to what extent, individual strains colonize and persist and how selective pressures define their genomic architecture. In this study, we combined shotgun sequencing of 1,203 stool samples from 26 mothers and their twins (52 infants), sampled from childbirth to 8 years after birth, with culture-enhanced, deep short-read and long-read stool sequencing from a subset of 10 twins (20 infants) to define transmission, persistence and evolutionary trajectories of gut species from infancy to middle childhood. We constructed 3,995 strain-resolved metagenome-assembled genomes across 399 taxa, and we found that 27.4% persist within individuals. We identified 726 strains shared within families, with Bacteroidales, Oscillospiraceae and Lachnospiraceae, but not Bifidobacteriaceae, vertically transferred. Lastly, we identified weaning as a critical inflection point that accelerates bacterial mutation rates and separates functional profiles of genes accruing mutations.
Collapse
Affiliation(s)
- Sanjam S Sawhney
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert Thänert
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Anna Thänert
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carla Hall-Moore
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - I Malick Ndao
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Bejan Mahmud
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Barbara B Warner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Phillip I Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gautam Dantas
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
53
|
Chu YL, Georgeson P, Clendenning M, Mahmood K, Walker R, Como J, Joseland S, Preston SG, Rice T, Lynch BM, Milne RL, Southey MC, Giles GG, Phipps AI, Hopper JL, Win AK, Rosty C, Macrae FA, Winship I, Jenkins MA, Buchanan DD, Joo JE. Intratumoural pks +Escherichia coli is associated with risk of metachronous colorectal cancer and adenoma development in people with Lynch syndrome. EBioMedicine 2025; 114:105661. [PMID: 40158390 PMCID: PMC11995779 DOI: 10.1016/j.ebiom.2025.105661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND The adverse gut microbiome may underlie the variability in risks of colorectal cancer (CRC) and metachronous CRC in people with Lynch syndrome (LS). The role of pks+/-Escherichia coli (pks+/-E. coli), Enterotoxigenic Bacteroides fragilis (ETBF), and Fusobacterium nucleatum (Fn) in CRCs and adenomas in people with LS is unknown. METHODS A total of 358 LS cases, including 386 CRCs, 90 adenomas, 195 normal colonic mucosa DNA from the Australasian Colon Cancer Family Registry were tested using multiplex TaqMan qPCR. Logistic regression was used to compare the intratumoural prevalence of each bacteria in Lynch CRCs with 1336 sporadic CRCs. Cox proportional-hazards regression estimated the associations of each bacteria with the risk of metachronous CRC and neoplasia. FINDINGS Pks+ E. coli (odds ratio [95% confidence interval] = 1.60 [1.08-2.35], P = 0.017), pks-E. coli (3.87 [2.58-5.80], P < 0.001) and Fn (19.47 [13.32-28.87], P < 0.001) were significantly enriched in LS CRCs when compared with sporadic CRCs. Pks+ E. coli in the initial CRC was associated with an increased risk of metachronous CRC (hazard ratio [95% confidence interval] = 2.32 [1.29-4.17], P = 0.005) and metachronous colorectal neoplasia (1.51 [1.02-2.23], P = 0.040) when compared with CRCs without pks+ E. coli. INTERPRETATION Pks+ E. coli, pks-E. coli, and Fn are enriched within LS CRCs, suggesting possible roles in CRC development in LS. Having intratumoural pks+ E. coli is associated with increased risk of metachronous CRC, suggesting that, if validated, people with LS might benefit from pks+ E. coli screening and eradication. FUNDING This work was funded by an NHMRC Investigator grant (GNT1194896) and a Cancer Australia/Cancer Council NSW co-funded grant (GNT2012914).
Collapse
Affiliation(s)
- Yen Lin Chu
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Peter Georgeson
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Mark Clendenning
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Khalid Mahmood
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Melbourne Bioinformatics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Romy Walker
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Julia Como
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Sharelle Joseland
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Susan G Preston
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Toni Rice
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Brigid M Lynch
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Melissa C Southey
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Amanda I Phipps
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Aung K Win
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Christophe Rosty
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Queensland, Brisbane, Queensland, Australia; Envoi Specialist Pathologists, Brisbane, Queensland, Australia
| | - Finlay A Macrae
- Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Parkville, Victoria, Australia; Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Melbourne, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Ingrid Winship
- Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Melbourne, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Mark A Jenkins
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Melbourne, Victoria, Australia
| | - Jihoon E Joo
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
54
|
Breideband L, Wächtershäuser KN, Sarkar R, Puspathasan M, Stelzer EH, Pampaloni F. Gravitational forces and matrix stiffness modulate the invasiveness of breast cancer cells in bioprinted spheroids. Mater Today Bio 2025; 31:101640. [PMID: 40124331 PMCID: PMC11930500 DOI: 10.1016/j.mtbio.2025.101640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/29/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
The progression of breast cancer is influenced by the stiffness of the extracellular matrix (ECM), which becomes stiffer as cancer advances due to increased collagen IV and laminin secretion by cancer-associated fibroblasts. Intriguingly, breast cancer cells cultivated in two-dimensions exhibit a less aggressive behavior when exposed to weightlessness, or microgravity conditions. This study aims to elucidate the interplay between matrix stiffness and microgravity on breast cancer progression. For this purpose, three-dimensional spheroids of breast cancer cell lines (MCF-7 and MDA-MB-231) were formed. These spheroids were subsequently bioprinted in hydrogels of varying stiffness, obtained by the mixing of gelatin methacrylate and poly(ethylene) glycol diacrylate mixed at different ratios. The constructs were printed with a custom stereolithography (SLA) bioprinter converted from a low-cost, commercially available 3D printer. These bioprinted structures, encapsulating breast cancer spheroids, were then placed in a clinostat (microgravity simulation device) for a duration of seven days. Comparative analyses were conducted between objects cultured under microgravity and standard earth gravity conditions. Protein expression was characterized through fluorescent microscopy, while gene expression of MCF-7 constructs was analyzed via RNA sequencing. Remarkably, the influence of a stiffer ECM on the protein and gene expression levels of breast cancer cells could be modulated and sometimes even reversed in microgravity conditions. The study's findings hold implications for refining therapeutic strategies for advanced breast cancer stages - an array of genes involved in reversing aggressive or even metastatic behavior might lead to the discovery of new compounds that could be used in a clinical setting.
Collapse
Affiliation(s)
- Louise Breideband
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Kaja Nicole Wächtershäuser
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Ryan Sarkar
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Melosha Puspathasan
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Ernst H.K. Stelzer
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Francesco Pampaloni
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| |
Collapse
|
55
|
Qin H, Zhong Y, Huang J, Miao Y, Du M, Huang K. TRIM56 Promotes White Adipose Tissue Browning to Attenuate Obesity by Degrading TLE3. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414073. [PMID: 39928840 PMCID: PMC11967773 DOI: 10.1002/advs.202414073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/28/2025] [Indexed: 02/12/2025]
Abstract
In mammals, the activation of thermogenic adipocytes, such as beige and brown adipocytes, can significantly increase overall energy expenditure, offering a promising strategy to combat metabolic diseases. Despite its considerable potential, the regulatory mechanisms governing this activation remain largely elusive. This study bridges this gap by elucidating that tripartite motif 56 (TRIM56), an E3 ubiquitin ligase, is upregulated in response to cold stimuli, thereby promoting the recruitment of beige adipocytes. Notably, the overexpression of TRIM56 in adipocytes is shown to help mice maintain a core temperature under cold conditions, as well as confer protection against diet-induced obesity. Mechanistically, TRIM56 facilitates the degradation of the transducin-like enhancer protein 3 (TLE3) protein by promoting its K48-linked ubiquitination, which subsequently triggers the activation of thermogenic genes in subcutaneousl white adipose tissue and improved the metabolic profiles. These findings unveil a novel function for TRIM56 in adipocyte browning, suggesting its potential as a therapeutic target for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Haojie Qin
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yi Zhong
- Department of Rheumatology and ImmunologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Jinhui Huang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yanli Miao
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Meng Du
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Kai Huang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular AgingHuazhong University of Science and TechnologyWuhan430022China
- Hubei Clinical Research Center of Metabolic and Cardiovascular DiseaseHuazhong University of Science and TechnologyWuhan430022China
| |
Collapse
|
56
|
Ruiz‐Malagón AJ, Rodríguez‐Sojo MJ, Redondo E, Rodríguez‐Cabezas ME, Gálvez J, Rodríguez‐Nogales A. Systematic review: The gut microbiota as a link between colorectal cancer and obesity. Obes Rev 2025; 26:e13872. [PMID: 39614602 PMCID: PMC11884970 DOI: 10.1111/obr.13872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 10/11/2024] [Accepted: 10/25/2024] [Indexed: 12/01/2024]
Abstract
Microbiome modulation is one of the novel strategies in medicine with the greatest future to improve the health of individuals and reduce the risk of different conditions, including metabolic, immune, inflammatory, and degenerative diseases, as well as cancer. Regarding the latter, many studies have reported the role of the gut microbiome in carcinogenesis, formation and progression of colorectal cancer (CRC), as well as its response to different systemic therapies. Likewise, obesity, one of the most important risk factors for CRC, is also well known for its association with gut dysbiosis. Moreover, obesity and CRC display, apart from microbial dysbiosis, chronic inflammation, which participates in their pathogenesis. Although human and murine studies demonstrate the significant impact of the microbiome in regulating energy metabolism and CRC development, little is understood about the contribution of the microbiome to the development of obesity-associated CRC. Therefore, this systematic review explores the evidence for microbiome changes associated with these conditions and hypothesizes that this may contribute to the pathogenesis of obesity-related CRC. Two databases were searched, and different studies on the relationship among obesity, intestinal microbiota and CRC in clinical and preclinical models were selected. Data extraction was carried out by two reviewers independently, and 101 studies were finally considered. Findings indicate the existence of a risk association between obesity and CRC derived from metabolic, immune, and microbial disorders.
Collapse
Affiliation(s)
- Antonio Jesús Ruiz‐Malagón
- Department of Pharmacology, Center for Biomedical Research (CIBM)University of GranadaGranadaSpain
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA)GranadaSpain
- Instituto de Investigación Biomédica de Málaga (IBIMA)MalgaSpain
| | - María Jesús Rodríguez‐Sojo
- Department of Pharmacology, Center for Biomedical Research (CIBM)University of GranadaGranadaSpain
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA)GranadaSpain
| | - Eduardo Redondo
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA)GranadaSpain
- Servicio de DigestivoHospital Universitario Virgen de las NievesGranadaSpain
| | - María Elena Rodríguez‐Cabezas
- Department of Pharmacology, Center for Biomedical Research (CIBM)University of GranadaGranadaSpain
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA)GranadaSpain
| | - Julio Gálvez
- Department of Pharmacology, Center for Biomedical Research (CIBM)University of GranadaGranadaSpain
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA)GranadaSpain
| | - Alba Rodríguez‐Nogales
- Department of Pharmacology, Center for Biomedical Research (CIBM)University of GranadaGranadaSpain
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA)GranadaSpain
| |
Collapse
|
57
|
Parekh Z, Xiao J, Mani A, Evans Q, Phung C, Barba HA, Xie B, Sidebottom AM, Sundararajan A, Lin H, Ramaswamy R, Dao D, Gonnah R, Yehia M, Hariprasad SM, D'Souza M, Sulakhe D, Chang EB, Skondra D. Fecal Microbial Profiles and Short-Chain Fatty Acid/Bile Acid Metabolomics in Patients With Age-Related Macular Degeneration: A Pilot Study. Invest Ophthalmol Vis Sci 2025; 66:21. [PMID: 40202735 PMCID: PMC11993127 DOI: 10.1167/iovs.66.4.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 02/25/2025] [Indexed: 04/10/2025] Open
Abstract
Purpose Age-related macular degeneration (AMD) is a multifactorial disease, and studies have implicated the role of gut microbiota in its pathogenesis. However, characterization of microbiome dysbiosis and associated microbial-derived metabolomic profiles across AMD stages remains unknown. In this pilot study, we explored how gut microbiome composition and gut-derived metabolites differ in AMD. Methods Our pilot study analyzed fasted stool samples that were collected from 22 patients at a tertiary academic center. Subjects were classified as control, intermediate AMD, or advanced AMD based on clinical presentation. 16S rRNA amplicon sequencing and standard chromatography-mass spectrometry methods were used to identify bacterial taxonomy composition and abundance of short-chain fatty acids (SCFAs) and bile acids (BAs), respectively. Genetic testing was used to investigate the frequency of 14 high-risk single nucleotide polymorphisms (SNPs) associated with AMD in the AMD cohort. Results Forty-three differentially abundant genera were present among the control, intermediate, and advanced groups. Taxa with known roles in immunologic pathways, such as Desulfovibrionales (q = 0.10) and Terrisporobacter (q = 1.16e-03), were in greater abundance in advanced AMD patients compared to intermediate. Advanced AMD patients had decreased abundance of 12 SCFAs, including acetate (P = 0.002), butyrate (P = 0.04), and propionate (P = 0.01), along with 12 BAs, including taurocholic acid (P = 0.02) and tauroursodeoxycholic acid (P = 0.04). Frequencies of high-risk SNPs were not significantly different between the intermediate and advanced AMD groups. Conclusions This pilot study identifies distinct gut microbiome compositions and metabolomic profiles associated with AMD and its stages, providing preliminary evidence of a potential link between gut microbiota and AMD pathogenesis. To validate these findings and elucidate the underlying mechanisms, future research with larger cohorts and more comprehensive sampling is strongly recommended.
Collapse
Affiliation(s)
- Zaid Parekh
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Jason Xiao
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Amir Mani
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Quadis Evans
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Christopher Phung
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Hugo A. Barba
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Bingqing Xie
- Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Ashley M. Sidebottom
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Anitha Sundararajan
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Huaiying Lin
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Ramanujam Ramaswamy
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - David Dao
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Reem Gonnah
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Madeleine Yehia
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Seenu M. Hariprasad
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Mark D'Souza
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Dinanath Sulakhe
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Eugene B. Chang
- Department of Medicine, The University of Chicago, Chicago, Illinois, United States
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Dimitra Skondra
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
58
|
Yan W, Wang S, Zhu L, Yu X, Li J. Targeted editing of CCL5 with CRISPR-Cas9 nanoparticles enhances breast cancer immunotherapy. Apoptosis 2025; 30:912-935. [PMID: 39870938 PMCID: PMC11947030 DOI: 10.1007/s10495-024-02032-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2024] [Indexed: 01/29/2025]
Abstract
Breast cancer remains one of the leading causes of cancer-related mortality among women worldwide. Immunotherapy, a promising therapeutic approach, often faces challenges due to the immunosuppressive tumor microenvironment. This study explores the innovative use of CRISPR-Cas9 technology in conjunction with FCPCV nanoparticles to target and edit the C-C Motif Chemokine Ligand 5 (CCL5) gene, aiming to improve the efficacy of breast cancer immunotherapy. Single-cell RNA sequencing (scRNA-seq) and TCGA-BRCA data identified CCL5 as a key immune-related gene in breast cancer. Using CRISPR-Cas9, sgRNA targeting CCL5 was designed and delivered to breast cancer cells and humanized mouse models via FCPCV nanoparticles. In vitro experiments demonstrated that FCPCV nanoparticles effectively silenced CCL5, enhanced CD8+ T cell activity, and increased the production of cytokines such as IFN-γ, TNF-α, and GZMB. In vivo studies revealed significant tumor suppression, improved immune microenvironment, and increased CD8+/CD4+ ratios in treated mice, without notable toxic side effects. These findings highlight the potential of CRISPR-Cas9 nanoparticle-mediated gene editing as a novel strategy for enhancing breast cancer immunotherapy, providing a new direction for personalized and effective cancer treatment.
Collapse
Affiliation(s)
- Wei Yan
- Department of Thoracic Oncology, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Key Laboratory of Oncology, Nanchang, 330029, China
| | - Shuo Wang
- Department of Thoracic Oncology, Ganzhou Cancer Hospital, Ganzhou Institute for Cancer Research, The Affiliated Cancer Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Lihui Zhu
- Department of Endoscopy Center, Jiangxi Provincial Children's Hospital, Nanchang, 330006, China
| | - Xinlin Yu
- Department of Medical Laboratory, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Key Laboratory of Oncology, No. 519 Beijing East Road, Nanchang, Jiangxi, 330029, China.
| | - Jianglong Li
- Department of Breast Cancer Surgery, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Key Laboratory of Oncology, No. 519 Beijing East Road, Nanchang, Jiangxi, 330029, China.
| |
Collapse
|
59
|
Xiong Q, Zhang Y, Zheng Y, Zhu Q. Regulation and application of m 6A modification in tumor immunity. SCIENCE CHINA. LIFE SCIENCES 2025; 68:974-993. [PMID: 39648245 DOI: 10.1007/s11427-024-2648-0] [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: 03/02/2024] [Accepted: 06/11/2024] [Indexed: 12/10/2024]
Abstract
The m6A modification is an RNA modification that impacts various processes of RNA molecules, including transcription, splicing, stability, and translation. Recently, researchers have discovered that the presence of m6A modification can influence the interaction between tumor cells and immune cells and also play a role in regulating the expression of immune response-related genes. Additionally, m6A modification is intricately involved in the regulation of tumor immune evasion and drug resistance. Specifically, certain tumor cells can manipulate the gene expression through m6A modification to evade immune system attacks. Therefore, it might be possible to enhance tumor immune surveillance and improve the effectiveness of immune-based therapies by manipulating m6A modification. This review systematically discusses the role of m6A modification in tumor immunity, specifically highlighting its regulation of immune cells and immune-related genes in tumor cells. Furthermore, we explore the potential of m6A modification inhibitors as anti-cancer therapies and the significance of m6A regulatory factors in predicting the efficacy of tumor immune therapy.
Collapse
Affiliation(s)
- Qunli Xiong
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yaguang Zhang
- Laboratory of Gastrointestinal Tumor Epigenetics and Genomics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Zheng
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
60
|
Rewell SSJ, Shad A, Chen L, Macowan M, Chu E, Gandasasmita N, Casillas-Espinosa PM, Li J, O'Brien TJ, Semple BD. A post-injury immune challenge with lipopolysaccharide following adult traumatic brain injury alters neuroinflammation and the gut microbiome acutely, but has little effect on chronic outcomes. Exp Neurol 2025; 386:115150. [PMID: 39842491 DOI: 10.1016/j.expneurol.2025.115150] [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: 09/29/2024] [Revised: 12/22/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
Patients with a traumatic brain injury (TBI) are susceptible to hospital-acquired infections, presenting a significant challenge to an already-compromised immune system. The consequences and mechanisms by which this dual insult worsens outcomes are poorly understood. This study aimed to explore how a systemic immune stimulus (lipopolysaccharide, LPS) influences outcomes following experimental TBI in young adult mice. Male and female C57Bl/6J mice underwent controlled cortical impact or sham surgery, followed by 1 mg/kg i.p. LPS or saline-vehicle at 4 days post-TBI, before behavioral assessment and tissue collection at 6 h, 24 h, 7 days or 6 months. LPS induced acute sickness behaviors including weight loss, transient hypoactivity, and increased anxiety-like behavior. Early systemic immune activation by LPS was confirmed by increased spleen weight and serum cytokines. In brain tissue, gene expression analysis revealed a time course of inflammatory immune activation in TBI or LPS-treated mice (e.g., IL-1β, IL-6, CCL2, TNFα), which was exacerbated in TBI + LPS mice. This group also presented with fecal microbiome dysbiosis at 24 h post-LPS, with reduced bacterial diversity and changes in the relative abundance of key bacterial genera associated with sub-acute neurobehavioral and immune changes. Chronically, TBI induced hyperactivity and cognitive deficits, brain atrophy, and increased seizure susceptibility, similarly in vehicle and LPS-treated groups. Together, findings suggest that an immune challenge with LPS early after TBI, akin to a hospital-acquired infection, alters the acute neuroinflammatory response to injury, but has no lasting effects. Future studies could consider more clinically-relevant models of infection to build upon these findings.
Collapse
Affiliation(s)
- Sarah S J Rewell
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Alfred Health, Prahran, VIC, Australia
| | - Ali Shad
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Alfred Health, Prahran, VIC, Australia
| | - Lingjun Chen
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Matthew Macowan
- Department of Immunology, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Erskine Chu
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Natasha Gandasasmita
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Alfred Health, Prahran, VIC, Australia
| | - Jian Li
- Department of Microbiology, Monash Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Alfred Health, Prahran, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, VIC, Australia
| | - Bridgette D Semple
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
61
|
Wu J, Feng H, Wang ZY, Li J. Factors Affecting Liver Function Abnormalities After Laparoscopic Esophageal Hiatal Hernia Repair. Surg Laparosc Endosc Percutan Tech 2025; 35:e1350. [PMID: 39632373 DOI: 10.1097/sle.0000000000001350] [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: 08/16/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE In this study, we investigated the factors related to abnormal liver function in patients undergoing laparoscopic esophageal hiatal hernia repair. METHODS The clinical data of 347 patients who underwent elective laparoscopic esophageal hiatal hernia repair at Beijing Chao-yang Hospital of Capital Medical University between January 2018 and November 2023 were retrospectively collected. The patients comprised 131 males and 216 females, ranging in age from 24 to 87 years, and were assessed using the ASA grading system between grades I and III. The patients were divided into 2 groups based on the presence or absence of liver function abnormalities on the first day after surgery: a normal liver function group (NLA group) and an abnormal liver function group (LA group). Patients with elevation in any of the following indicators were included in the LA group: alanine aminotransferase >40 U/L, glutamine aminotransferase >40 U/L, γ-glutamyltransferase >49 U/L, alkaline phosphatase >135 U/L, total bilirubin >17.1 μmol/L, or direct bilirubin >6.8 μmol/L. The clinical data of the 2 groups of patients were compared, and only the indicators with a P -value <0.15 were included in a binary logistic regression model analysis. RESULTS There were 238 patients (68.6%) who developed liver function abnormalities on the first postoperative day. In comparison to the NLA group, the LA group had a significantly higher proportion of patients with esophageal hiatal hernia type II, type III, and type IV, hypotension, and high P ET CO 2 . Furthermore, the LA group had a significantly lower proportion of patients receiving blood transfusions. The maximum length and maximum cross-sectional area of the esophageal hiatal hernia were also significantly larger in the LA group. In addition, the operation time was significantly longer in the LA group. (all P -values are <0.15). The binary logistic regression analysis revealed that prolonged operation time (OR=1.017, 95% CI: 1.007-1.028) was the only risk factor associated with postoperative liver function abnormalities. CONCLUSIONS The sole risk factor for postoperative liver function abnormalities was prolonged surgical time.
Collapse
Affiliation(s)
- Jin Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | | | | | | |
Collapse
|
62
|
Murgiano M, Bartocci B, Puca P, di Vincenzo F, Del Gaudio A, Papa A, Cammarota G, Gasbarrini A, Scaldaferri F, Lopetuso LR. Gut Microbiota Modulation in IBD: From the Old Paradigm to Revolutionary Tools. Int J Mol Sci 2025; 26:3059. [PMID: 40243712 PMCID: PMC11988433 DOI: 10.3390/ijms26073059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/18/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders primarily comprising two main conditions: ulcerative colitis and Crohn's disease. The gut microbiota's role in driving inflammation in IBD has garnered significant attention, yet the precise mechanisms through which the microbiota influences IBD pathogenesis remain largely unclear. Given the limited therapeutic options for IBD, alternative microbiota-targeted therapies-including prebiotics, probiotics, postbiotics, and symbiotics-have been proposed. While these approaches have shown promising results, microbiota modulation is still mainly considered an adjunct therapy to conventional treatments, with a demonstrated impact on patients' quality of life. Fecal microbiota transplantation (FMT), already approved for treating Clostridioides difficile infection, represents the first in a series of innovative microbiota-based therapies under investigation. Microbial biotherapeutics are emerging as personalized and cutting-edge tools for IBD management, encompassing next-generation probiotics, bacterial consortia, bacteriophages, engineered probiotics, direct metabolic pathway modulation, and nanotherapeutics. This review explores microbial modulation as a therapeutic strategy for IBDs, highlighting current approaches and examining promising tools under development to better understand their potential clinical applications in managing intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Marco Murgiano
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Bianca Bartocci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Pierluigi Puca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Federica di Vincenzo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Angelo Del Gaudio
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Alfredo Papa
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Franco Scaldaferri
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Loris Riccardo Lopetuso
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Dipartimento di Scienze della Vita, della Salute e delle Professioni Sanitarie, Università degli Studi Link, 00165 Rome, Italy
| |
Collapse
|
63
|
Yan Q, Huang L, Li S, Zhang Y, Guo R, Zhang P, Lei Z, Lv Q, Chen F, Li Z, Meng J, Li J, Wang G, Chen C, Ullah H, Cheng L, Fan S, You W, Zhang Y, Ma J, Sha S, Sun W. The Chinese gut virus catalogue reveals gut virome diversity and disease-related viral signatures. Genome Med 2025; 17:30. [PMID: 40140988 PMCID: PMC11938785 DOI: 10.1186/s13073-025-01460-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND The gut viral community has been increasingly recognized for its role in human physiology and health; however, our understanding of its genetic makeup, functional potential, and disease associations remains incomplete. METHODS In this study, we collected 11,286 bulk or viral metagenomes from fecal samples across large-scale Chinese populations to establish a Chinese Gut Virus Catalogue (cnGVC) using a de novo virus identification approach. We then examined the diversity and compositional patterns of the gut virome in relation to common diseases by analyzing 6311 bulk metagenomes representing 28 disease or unhealthy states. RESULTS The cnGVC contains 93,462 nonredundant viral genomes, with over 70% of these being novel viruses not included in existing gut viral databases. This resource enabled us to characterize the functional diversity and specificity of the gut virome. Using cnGVC, we profiled the gut virome in large-scale populations, assessed sex- and age-related variations, and identified 4238 universal viral signatures of diseases. A random forest classifier based on these signatures achieved high accuracy in distinguishing diseased individuals from controls (AUC = 0.698) and high-risk patients from controls (AUC = 0.761), and its predictive ability was also validated in external cohorts. CONCLUSIONS Our resources and findings significantly expand the current understanding of the human gut virome and provide a comprehensive view of the associations between gut viruses and common diseases. This will pave the way for novel strategies in the treatment and prevention of these diseases.
Collapse
Affiliation(s)
- Qiulong Yan
- The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, 510900, China.
- Department of Microbiology, Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| | - Liansha Huang
- Department of Reproductive Health, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518101, China
| | - Shenghui Li
- Puensum Genetech Institute, Wuhan, 430076, China
| | - Yue Zhang
- Puensum Genetech Institute, Wuhan, 430076, China
| | - Ruochun Guo
- The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, 510900, China
| | - Pan Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Zhixin Lei
- School of Chemistry, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Disease, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, China.
| | - Qingbo Lv
- Puensum Genetech Institute, Wuhan, 430076, China
| | - Fang Chen
- The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, 510900, China
- Department of Microbiology, Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | | | - Jinxin Meng
- Puensum Genetech Institute, Wuhan, 430076, China
| | - Jing Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, 100044, China
| | - Guangyang Wang
- The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, 510900, China
- Department Pathology, Dalian Municipal Central Hospital, Dalian, 116033, China
| | - Changming Chen
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Hayan Ullah
- Department of Microbiology, Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Lin Cheng
- Department of Microbiology, Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Shao Fan
- Department of Microbiology, Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Wei You
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Yan Zhang
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Jie Ma
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Shanshan Sha
- Department of Microbiology, Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| | - Wen Sun
- Centre for Translational Medicine, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518101, China.
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
64
|
Song Y, An Q, Chen S, Dai H, Ma S, Wu C, Lyu Y, Shen J, Krüger-Haker H, Schwarz S, Wang L, Wang Y, Xia Z. Antimicrobial resistance of pet-derived bacteria in China, 2000-2020. Antimicrob Agents Chemother 2025; 69:e0165724. [PMID: 40135877 PMCID: PMC12057338 DOI: 10.1128/aac.01657-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025] Open
Abstract
With the rapid growth of the pet industry in China, bacterial infectious diseases in pets have increased, highlighting the need to monitor antimicrobial resistance (AMR) in pet-derived bacteria to improve the diagnosis and treatment. Before the establishment of the China Antimicrobial Resistance Surveillance Network for Pets (CARPet) in 2021, a comprehensive analysis of such data in China was lacking. Our review of 38 point-prevalence surveys conducted between 2000 and 2020 revealed increasing trends in AMR among pet-derived Escherichia coli, Klebsiella pneumoniae, Staphylococcus spp., Enterococcus spp., and other bacterial pathogens in China. Notable resistance to β-lactams and fluoroquinolones, which are largely used in both pets and livestock animals, was observed. For example, resistance rates for ampicillin and ciprofloxacin in E. coli frequently exceeded 50.0%, with up to 41.3% of the isolates producing extended-spectrum β-lactamases. The emergence of carbapenem-resistant K. pneumoniae and E. coli, carrying blaNDM and blaOXA genes, highlighted the need for vigilant monitoring. The detection rate of SCCmec (Staphylococcal Cassette Chromosome mec), a genetic element associated with methicillin resistance, in Staphylococcus pseudintermedius isolated from pets in China was found to be over 40.0%. The resistance rate of E. faecalis to vancomycin was 2.1% (5/223) in East China, which was higher than the detection rate of human-derived vancomycin-resistant Enterococcus (0.1%, 12/11,215). Establishing the national AMR surveillance network CARPet was crucial, focusing on representative cities, diverse clinical samples, and including both commonly used antimicrobial agents in veterinary practice and critically important antimicrobial agents for human medicine, such as carbapenems, tigecycline, and vancomycin.
Collapse
Affiliation(s)
- Yu Song
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qi An
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Siyu Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hegen Dai
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shizhen Ma
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Congming Wu
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yanli Lyu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Henrike Krüger-Haker
- Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, Berlin, Germany
| | - Stefan Schwarz
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, Berlin, Germany
| | - Lu Wang
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yang Wang
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhaofei Xia
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
65
|
Liu Y, Li X, Chen Y, Yao Q, Zhou J, Wang X, Meng Q, Ji J, Yu Z, Chen X. Fecal microbiota transplantation: application scenarios, efficacy prediction, and factors impacting donor-recipient interplay. Front Microbiol 2025; 16:1556827. [PMID: 40201444 PMCID: PMC11975908 DOI: 10.3389/fmicb.2025.1556827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Fecal microbiota transplantation (FMT) represents a therapeutic approach that directly regulates the gut microbiota of recipients, normalizes its composition and reaping therapeutic rewards. Currently, in addition to its general application in treating Clostridium difficile (C. difficile) infection (CDI), FMT treatment has also been extended to the fields of other gastrointestinal diseases, infections, gut-liver or gut-brain axis disorders, metabolic diseases and cancer, etc. Prior to FMT, rigorous donor screening is essential to reduce the occurrence of adverse events. In addition, it is imperative to evaluate whether the recipient can safely and effectively undergo FMT treatment. However, the efficacy of FMT is influenced by the complex interactions between the gut microbiota of donor and recipient, the degree of donor microbiota engraftment is not necessarily positively related with the success rate of FMT. Furthermore, an increasing number of novel factors affecting FMT outcomes are being identified in recent clinical trials and animal experiments, broadening our understanding of FMT treatment. This article provides a comprehensive review of the application scenarios of FMT, the factors influencing the safety and efficacy of FMT from the aspects of both the donors and the recipients, and summarizes how these emerging novel regulatory factors can be combined to predict the clinical outcomes of patients undergoing FMT.
Collapse
Affiliation(s)
- Yaxin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinru Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuchao Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qinyan Yao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinjie Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoxuan Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qingguo Meng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiaxuan Ji
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Zihan Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
66
|
Jiang Z, Li P, Qiu K, Liao Y, Chen X, Xuan J, Wang F, Ma H, Wang Y, Zhu M. Proteus mirabilis exacerbates ulcerative colitis by inhibiting mucin production. Front Microbiol 2025; 16:1556953. [PMID: 40201443 PMCID: PMC11975560 DOI: 10.3389/fmicb.2025.1556953] [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: 01/07/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Introduction Ulcerative colitis (UC) is characterized by chronic inflammation and ulceration in colonic mucosa, accompanied by a defective epithelial barrier. Proteus mirabilis (P. mirabilis) bacterium is a putative intestinal pathogen with invasive ability, yet its role in UC inflammation and gut barrier disruption is unclear. This study aims to investigate its epidemiological presence, pathogenic roles and preventive strategy during UC inflammation. Method P. mirabilis culture and PCR amplification of the P. mirabilis-specific ureR gene were used to detect fecal P. mirabilis and determine its prevalence in UC and control stool specimens. P. mirabilis isolated from UC stool specimens was gavaged into dextran sulfate sodium (DSS)-treated mice. Inflammation and the mucus layer of colons were assessed through histological examination and cytokine quantification. Bacteriophages were screened and used to eliminate P. mirabilis in colitis animals. Results and discussion The fecal P. mirabilis bacteria were detected by PCR amplification of P. mirabilis-specific ureR gene. Of 41 UC patients, 65.9% patients were P. mirabilis positive, which was significantly higher than the controls. Administration of P. mirabilis aggravated DSS-induced colitis symptom and mucosal inflammation in mice. Interestingly, the colonic mucus layer, an essential component of the epithelial barrier, of the animals was dramatically disrupted, which was consistent with the alteration of human UC colon. The disrupted mucus layer was mediated by the down-regulation of IL-18 in intestinal epithelium. Importantly, a bacteriophage cocktail targeting P. mirabilis could restore the mucus barrier and alleviate the enteric inflammation. Thus, our results suggest that P. mirabilis is a UC pathobiont bacterium, which exacerbates the severity of UC inflammation owing to down-regulation of mucin production and IL-18 expression. Bacteriophage-mediated elimination of P. mirabilis may be effective in limiting UC inflammation.
Collapse
Affiliation(s)
- Zhihui Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| | - Pengpeng Li
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| | - Kehui Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| | - Yang Liao
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| | - Xin Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| | - Ji Xuan
- Department of Gastroenterology, Jinling Hospital, The Medical School of Nanjing University, Nanjing, China
| | - Fangyu Wang
- Department of Gastroenterology, Jinling Hospital, The Medical School of Nanjing University, Nanjing, China
| | - Hongfeng Ma
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
- Department of Rehabilitation Medicine, Huzhou Rehabilitation Hospital, Huzhou, China
| | - Ye Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| | - Minsheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
67
|
Zhang Y, Lin H, Xiong Y, Zhang Z, Zeng L, Liu Z. Fu Brick Tea Protects the Intestinal Barrier and Ameliorates Colitis in Mice by Regulating Gut Microbiota. Foods 2025; 14:1122. [PMID: 40238292 PMCID: PMC11989102 DOI: 10.3390/foods14071122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/16/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Ulcerative colitis (UC) pathogenesis is strongly linked to gut microbiota dysbiosis and compromised intestinal barrier integrity. Emerging evidence suggests that targeted dietary interventions may restore microbial homeostasis and ameliorate colitis progression. In this study, we evaluated the therapeutic potential of Fu Brick tea (FBT) using a dextran sulfate sodium (DSS)-induced murine colitis model. The results indicated that oral administration of FBT extract significantly improved the disease index, reduced inflammatory response, protected intestinal barrier protein (e.g., ZO-1), and maintained intestinal structure integrity. Furthermore, FBT intake increased the diversity of gut microbiota, promoted the growth of beneficial bacteria (e.g., Akkermansia), inhibited the proliferation of harmful bacteria (e.g., Desulfovibrioceae, Escherichia, and Helicobacter), restored intestinal homeostasis, and alleviated colitis symptoms including diarrhea. These findings position FBT as a promising nutraceutical candidate for UC management via multi-target modulation of mucosal immunity and microbial ecology.
Collapse
Affiliation(s)
- Yangbo Zhang
- School of Pharmacy, Shaoyang University, Shaoyang 422000, China;
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; (H.L.); (Y.X.)
| | - Haiyan Lin
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; (H.L.); (Y.X.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Yifan Xiong
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; (H.L.); (Y.X.)
| | - Zhixu Zhang
- Yuelushan Laboratory, Changsha 410128, China
| | - Li Zeng
- School of Pharmacy, Shaoyang University, Shaoyang 422000, China;
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; (H.L.); (Y.X.)
- Yuelushan Laboratory, Changsha 410128, China
| |
Collapse
|
68
|
Pueschel L, Wedemeyer H, Lenzen H, Wiestler M. Sex Differences Outweigh Dietary Factors in Food-Related Quality of Life in Patients with Inflammatory Bowel Disease. Nutrients 2025; 17:1114. [PMID: 40218872 PMCID: PMC11990271 DOI: 10.3390/nu17071114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), consists of chronic gastrointestinal inflammation, with nutrition playing a significant role in its development. IBD patients often face dietary challenges affecting their quality of life (QoL), yet research on food-related QoL (FR-QoL) and sex-specific differences is limited. It was hypothesized that dietary patterns and choices impact food-related quality of life in IBD and that these effects vary by sex. The objective of this analysis was, therefore, to evaluate the impact of diet on food-related quality of life for men and women with IBD, respectively. METHODS A monocentric, cross-sectional study at a tertiary referral center analyzed the food-related quality of life in 117 women and 116 men with IBD, with a particular focus on dietary choices and patterns. To achieve this, multiple assessment tools, including the German version of the IBD-specific Questionnaire for Food-Related Quality of Life (FR-QoL-29-German) and a validated Food Frequency Questionnaire (FFQ) for dietary behavior, were used. Clinical indices (Harvey-Bradshaw Index (HBI); Partial Mayo Score (PMS)) and biochemical markers (C-reactive protein; fecal calprotectin) were evaluated. RESULTS The FR-QoL-29-German sum score differed significantly between the sexes (p = 0.034; g = -0.3), with men showing a higher mean score. Distinct dietary patterns showed little correlation with FR-QoL for both sexes, except for a significant inverse correlation between FR-QoL and sQ-HPF scores for men (p = 0.021; r = -0.214) but not for women (p = 0.897; r = -0.012). In a logistic regression analysis that was adjusted for confounding, the impact of IBD-specific and diet-related factors on FR-QoL was assessed, and disease entity was identified as a significant influencing factor for men but not for women. In women, older age and lower body weight were associated with higher FR-QoL. CONCLUSIONS The findings of this study indicate that dietary choices and patterns do not exhibit uniform associations with IBD-related quality of life. In addition, sex differences have been identified as a substantial factor in IBD food-related quality of life.
Collapse
Affiliation(s)
- Lea Pueschel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Henrike Lenzen
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Department of Gastroenterology, Hepatology, Interventional Endoscopy and Diabetology, Academic Teaching Hospital Braunschweig, 38126 Braunschweig, Germany
| | - Miriam Wiestler
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- PRACTIS Clinician Scientist Program, Dean’s Office for Academic Career Development, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
69
|
Tartamus (Tita) GV, Serban DE, Fogas CR, Tantau MV. Pediatric Inflammatory Bowel Disease in Romania: The First Epidemiological Study of the North-West Region (2000-2020). CHILDREN (BASEL, SWITZERLAND) 2025; 12:403. [PMID: 40310072 PMCID: PMC12025974 DOI: 10.3390/children12040403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 05/02/2025]
Abstract
Background: Inflammatory bowel disease (IBD) represents a group of disorders with increasing incidence in the pediatric population worldwide. There are limited data on pediatric IBD (pIBD) epidemiology in Eastern Europe and none so far from Romania. The aim of our study was to evaluate incidence rates and time trends for pIBD in the north-west region of Romania and to compare them with results from studies on the same topic published worldwide. Methods: We performed a retrospective study of patients under 18 years of age diagnosed with pIBD in the time frame between 1 January 2000 and 31 December 2020 at the Emergency Clinical Hospital for Children in Cluj-Napoca. Age-adjusted incidence rates, annual percentage change (APC), average annual percentage chance (AAPC) and their corresponding 95% confidence intervals (CIs) were calculated for pIBD, Crohn's disease (CD) and ulcerative colitis (UC). Temporal trends were plotted using Joinpoint regression. Results: Ninety-four patients were identified. For the entire studied period, the incidence rate for pIBD was 0.79/105 (±0.74), 0.4/105 for CD (±0.42) and 0.34/105 for UC (±0.4). Time trends for incidence were positive, but statistical significance was found only for pIBD and CD. The APC observed for pIBD, CD and UC was 12 (95% CI: 6.5-17.7), 13.1 (95% CI: 8-18.5) and 5.67 (95% CI: 1.5-13.4), respectively. Comparison to other similar studies placed Romania among the countries with the lowest incidence of pIBD. Conclusions: Although pIBD incidence in our region appears to be low, there has been an important and significant increase in the incidence of overall pIBD and especially CD.
Collapse
Affiliation(s)
- Georgia Valentina Tartamus (Tita)
- 3rd Medical Discipline, Department of Internal Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (G.V.T.); (C.R.F.)
| | - Daniela Elena Serban
- Department of Mother and Child, 2nd Clinic of Pediatrics, Emergency Clinical Hospital for Children, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400177 Cluj-Napoca, Romania
| | - Cristina Rebeca Fogas
- 3rd Medical Discipline, Department of Internal Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (G.V.T.); (C.R.F.)
| | - Marcel Vasile Tantau
- Department of Internal Medicine and Gastroenterology, “Prof. Dr. Octavian Fodor”, Regional Institute of Gastroenterology and Hepatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania;
| |
Collapse
|
70
|
Bilal H, Ullah S, Halim SA, Khan M, Avula SK, Alam A, Zayed ES, El-Ghaiesh SH, Ogaly HA, Shah Z, Khan A, Al-Harrasi A. Design and synthesis of terephthalic dihydrazide analogues as dual inhibitors of glycation and urease. RSC Adv 2025; 15:9510-9520. [PMID: 40161525 PMCID: PMC11951093 DOI: 10.1039/d5ra00459d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
The overexpression of urease is the root cause of peptic ulcers and gastritis. Therefore, introducing new inhibitors against urease is a possible therapeutic approach to overcoming the pathogenesis; for instance, limiting the risk of development of urinary calculi. Moreover, glycation is the leading cause of several complications. Thus, in this study, we synthesized novel terephthalic dihydrazide analogues and evaluated their biological importance. These terephthalic dihydrazide analogues were characterized using advanced spectroscopic techniques, such as 1H NMR, 13C NMR, 19F NMR and HRMS (ESI+), and FT-IR. Fortunately, 6 of the 11 synthesized compounds exhibited urease inhibitory capability, and 8 compounds exhibited anti-glycation capability. Compounds 13-14, 20 and 23 showed significant urease inhibition with IC50 values of 63.12 ± 0.28, 65.71 ± 0.40, 49.2 ± 0.49 and 51.45 ± 0.39 μM, respectively. Meanwhile, they exhibited potent anti-glycation activity with IC50 values of 67.53 ± 0.46, 68.06 ± 0.43, 48.32 ± 0.42 and 54.36 ± 0.40 μM, respectively. Molecular docking of active urease inhibitors showed their good binding at the entrance of the active site and good correlation with our in vitro results.
Collapse
Affiliation(s)
- Hazrat Bilal
- Department of Chemistry, Bacha Khan University Charsadda Charsadda-24420 Khyber Pakhtunkhwa Pakistan
- Department of Chemistry, Government Postgraduate College Dargai Malakand Pakistan
| | - Saeed Ullah
- Natural and Medical Sciences Research Center, University of Nizwa PO Box 33, 616 Birkat Al Mauz Nizwa Sultanate of Oman +968-98957352 +968 25446328
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa PO Box 33, 616 Birkat Al Mauz Nizwa Sultanate of Oman +968-98957352 +968 25446328
| | - Momin Khan
- Department of Chemistry, Abdul Wali Khan University Mardan Mardan 23200 Pakistan
| | - Satya Kumar Avula
- Natural and Medical Sciences Research Center, University of Nizwa PO Box 33, 616 Birkat Al Mauz Nizwa Sultanate of Oman +968-98957352 +968 25446328
| | - Aftab Alam
- Department of Biochemistry, Abdul Wali Khan University Mardan Mardan 23200 Pakistan
| | - Eman Serry Zayed
- Department of Clinical Biochemistry, Faculty of Medicine, University of Tabuk Tabuk 71491 Saudi Arabia
| | - Sabah H El-Ghaiesh
- Department of Pharmacology, Faculty of Medicine, University of Tabuk Tabuk 71491 Saudi Arabia
- Department of Pharmacology, Faculty of Medicine, Tanta University Tanta Egypt
| | - Hanan A Ogaly
- Chemistry Department, College of Science, King Khalid University Abha 61421 Saudi Arabia
| | - Zarbad Shah
- Department of Chemistry, Bacha Khan University Charsadda Charsadda-24420 Khyber Pakhtunkhwa Pakistan
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa PO Box 33, 616 Birkat Al Mauz Nizwa Sultanate of Oman +968-98957352 +968 25446328
- Department of Chemical and Biological Engineering, College of Engineering, Korea University Seongbuk-gu 02841 Republic of Korea
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa PO Box 33, 616 Birkat Al Mauz Nizwa Sultanate of Oman +968-98957352 +968 25446328
| |
Collapse
|
71
|
Li Y, Xiao P, Boadu F, Goldkamp AK, Nirgude S, Cheng J, Hagen DE, Kalish JM, Rivera RM. Beckwith-Wiedemann syndrome and large offspring syndrome involve alterations in methylome, transcriptome, and chromatin configuration. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2023.12.14.23299981. [PMID: 38168424 PMCID: PMC10760283 DOI: 10.1101/2023.12.14.23299981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Beckwith-Wiedemann Syndrome (BWS) is the most common epigenetic overgrowth syndrome, caused by epigenetic alterations on chromosome 11p15. In ∼50% of patients with BWS, the imprinted region KvDMR1 (IC2) is hypomethylated. Nearly all children with BWS develop organ overgrowth and up to 28% develop cancer during childhood. The global epigenetic alterations beyond the 11p15 region in BWS are not currently known. Uncovering these alterations at the methylome, transcriptome, and chromatin architecture levels are necessary steps to improve the diagnosis and understanding of patients with BWS. Here we characterized the complete epigenetic profiles of BWS IC2 individuals together with the animal model of BWS, bovine large offspring syndrome (LOS). A novel finding of this research is the identification of two molecular subgroups of BWS IC2 individuals. Genome-wide alternations were detected for DNA methylation, transcript abundance, alternative splicing events of RNA, chromosome compartments, and topologically associating domains (TADs) in BWS and LOS, with shared alterations identified between species. Altered chromosome compartments and TADs were correlated with differentially expressed genes in BWS and LOS. Together, we highlight genes and genomic regions that have the potential to serve as targets for biomarker development to improve current molecular diagnostic methodologies for BWS.
Collapse
|
72
|
Rozera T, Pasolli E, Segata N, Ianiro G. Machine Learning and Artificial Intelligence in the Multi-Omics Approach to Gut Microbiota. Gastroenterology 2025:S0016-5085(25)00526-8. [PMID: 40118220 DOI: 10.1053/j.gastro.2025.02.035] [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: 12/10/2024] [Revised: 01/26/2025] [Accepted: 02/10/2025] [Indexed: 03/23/2025]
Abstract
The gut microbiome is involved in human health and disease, and its comprehensive understanding is necessary to exploit it as a diagnostic or therapeutic tool. Multi-omics approaches, including metagenomics, metatranscriptomics, metabolomics, and metaproteomics, enable depiction of the gut microbial ecosystem's complexity. However, these tools generate a large data stream in which integration is needed to produce clinically useful readouts, but, in turn, might be difficult to carry out with conventional statistical methods. Artificial intelligence and machine learning have been increasingly applied to multi-omics datasets in several conditions associated with microbiome disruption, from chronic disorders to cancer. Such tools have potential for clinical implementation, including discovery of microbial biomarkers for disease classification or prediction, prediction of response to specific treatments, and fine-tuning of microbiome-modulating therapies. The state of the art, potential, and limits, of artificial intelligence and machine learning in the multi-omics approach to gut microbiome are discussed.
Collapse
Affiliation(s)
- Tommaso Rozera
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, L'Unità Operativa Complessa Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy; Department of Medical and Surgical Sciences, L'Unità Operativa Complessa Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Edoardo Pasolli
- University of Naples Federico II, Department of Agricultural Sciences, Piazza Carlo di Borbone 1, Portici, Italy
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy; Department of Experimental Oncology, European Institute of Oncology Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, L'Unità Operativa Complessa Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy; Department of Medical and Surgical Sciences, L'Unità Operativa Complessa Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.
| |
Collapse
|
73
|
Redgwell TA, Thorsen J, Petit MA, Deng L, Vestergaard G, Russel J, Chawes B, Bønnelykke K, Bisgaard H, Nielsen DS, Sørensen S, Stokholm J, Shah SA. Prophages in the infant gut are pervasively induced and may modulate the functionality of their hosts. NPJ Biofilms Microbiomes 2025; 11:46. [PMID: 40108202 PMCID: PMC11923282 DOI: 10.1038/s41522-025-00674-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025] Open
Abstract
Gut microbiome (GM) composition and function is pivotal for human health and disease, of which the virome's importance is increasingly recognised. However, prophages and their induction patterns in the infant gut remain understudied. Here, we identified 10645 putative prophages in 662 metagenomes from 1-year-old children in the COPSAC2010 mother-child cohort and investigated their potential functions. No core provirome was found as the most prevalent vOTU was identified in only ~70% of the samples. The most dominant cluster of vOTUs in the cohort was related to Bacteroides phage Hanky p00', and it carried both diversity generating retroelements and genes involved in capsular polysaccharide synthesis. Paired analysis of viromes and metagenomes from the same samples revealed that most prophages within the infant gut were induced and that induction was unaffected by a range of environmental perturbers. In summary, prophages are major components of the infant gut that may have far reaching influences on the microbiome and its host.
Collapse
Affiliation(s)
- Tamsin A Redgwell
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark
| | - Jonathan Thorsen
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie-Agnès Petit
- Micalis institute, INRAE, Agroparistech, Université Paris-Saclay, Jouy en Josas, France
| | - Ling Deng
- Section of Food Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26, 1958, Frederiksberg C, Denmark
| | - Gisle Vestergaard
- Technical University of Denmark, Section of Bioinformatics, Department of Health Technology, 2800 Kgs, Lyngby, Denmark
| | - Jakob Russel
- Department of Biology, Section of Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Bo Chawes
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark
| | - Hans Bisgaard
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark
| | - Dennis S Nielsen
- Section of Food Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26, 1958, Frederiksberg C, Denmark
| | - Søren Sørensen
- Department of Biology, Section of Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark
- Section of Food Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26, 1958, Frederiksberg C, Denmark
| | - Shiraz A Shah
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Ledreborg Allé 34, DK-2820, Gentofte, Denmark.
| |
Collapse
|
74
|
Sheikh A, Curran MA. The influence of the microbiome on radiotherapy and DNA damage responses. Front Oncol 2025; 15:1552750. [PMID: 40165887 PMCID: PMC11955455 DOI: 10.3389/fonc.2025.1552750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent cancers in terms of diagnosis and mortality. Radiotherapy (RT) remains a mainstay of CRC therapy. As RT relies on DNA damage to promote tumor cell death, the activity of cellular DNA damage repair pathways can modulate cancer sensitivity to therapy. The gut microbiome has been shown to influence intestinal health and is independently associated with CRC development, treatment responses and outcomes. The microbiome can also modulate responses to CRC RT through various mechanisms such as community structure, toxins and metabolites. In this review we explore the use of RT in the treatment of CRC and the molecular factors that influence treatment outcomes. We also discuss how the microbiome can promote radiosensitivity versus radioprotection to modulate RT outcomes in CRC. Understanding the molecular interaction between the microbiome and DNA repair pathways can assist with predicting responses to RT. Once described, these connections between the microbiome and RT response can also be used to identify actionable targets for therapeutic development.
Collapse
Affiliation(s)
- Aadil Sheikh
- Department of Medical Education, Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| | - Michael A. Curran
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
75
|
Roy B, Cao K, Singh CO, Fang X, Yang H, Wei D. Advances in gut microbiota-related treatment strategies for managing colorectal cancer in humans. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0263. [PMID: 40072039 PMCID: PMC11899591 DOI: 10.20892/j.issn.2095-3941.2024.0263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/15/2025] [Indexed: 03/15/2025] Open
Abstract
Colorectal cancer (CRC) is a major contributor to global cancer-related mortality with increasing incidence rates in both developed and developing regions. Therefore, CRC presents a significant challenge to global health. The development of innovative tools for enhancing early CRC screening and diagnosis, along with novel treatments and therapies for improved management, remains an urgent necessity. CRC is intricately associated with the gut microbiota, which is integral to food digestion, nutrient generation, drug metabolism, metabolite production, immune enhancement, endocrine regulation, neurogenesis modulation, and the maintenance of physiologic and psychological equilibrium. Dysbiosis or imbalances in the gut microbiome have been implicated in various disorders, including CRC. Emerging evidence highlights the critical role of the gut microbiome in CRC pathogenesis and treatment, which presents potential opportunities for early detection and diagnosis. Despite substantial advances in understanding the relationship between the gut microbiota and CRC, significant challenges persist. Gaining a deeper and more detailed understanding of the interactions between the human microbiota and cancer is essential to fully realize the potential of the microbiota in cancer management. Unlike genetic factors, the gut microbiome is subject to modification, offering a promising avenue for the development of CRC treatments and drug discovery. This review provides an overview of the interactions between the human gut microbiome and CRC, while examining prospects for precision management of CRC.
Collapse
Affiliation(s)
- Bhaskar Roy
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Science, Hangzhou 310022, China
| | - Kunfeng Cao
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Science, Hangzhou 310022, China
- BGI Research, Shenzhen 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | | | - Huanming Yang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Science, Hangzhou 310022, China
- BGI Research, Shenzhen 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- James D. Watson Institute of Genome Sciences, Hangzhou 310029, China
| | - Dong Wei
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Science, Hangzhou 310022, China
- BGI Research, Shenzhen 518083, China
- Clin Lab, BGI Genomics, Beijing 100000, China
| |
Collapse
|
76
|
Sharma B, Agriantonis G, Twelker K, Ebelle D, Kiernan S, Siddiqui M, Soni A, Cheerasarn S, Simon W, Jiang W, Cardona A, Chapelet J, Agathis AZ, Gamboa A, Dave J, Mestre J, Bhatia ND, Shaefee Z, Whittington J. Gut Microbiota Serves as a Crucial Independent Biomarker in Inflammatory Bowel Disease (IBD). Int J Mol Sci 2025; 26:2503. [PMID: 40141145 PMCID: PMC11942158 DOI: 10.3390/ijms26062503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD), ulcerative colitis (UC), and IBD unclassified (IBD-U), is a complex intestinal disorder influenced by genetic, environmental, and microbial factors. Recent evidence highlights the gut microbiota as a pivotal biomarker and modulator in IBD pathogenesis. Dysbiosis, characterized by reduced microbial diversity and altered composition, is a hallmark of IBD. A consistent decrease in anti-inflammatory bacteria, such as Faecalibacterium prausnitzii, and an increase in pro-inflammatory species, including Escherichia coli, have been observed. Metabolomic studies reveal decreased short-chain fatty acids (SCFAs) and secondary bile acids, critical for gut homeostasis, alongside elevated pro-inflammatory metabolites. The gut microbiota interacts with host immune pathways, influencing morphogens, glycosylation, and podoplanin (PDPN) expression. The disruption of glycosylation impairs mucosal barriers, while aberrant PDPN activity exacerbates inflammation. Additionally, microbial alterations contribute to oxidative stress, further destabilizing intestinal barriers. These molecular and cellular disruptions underscore the role of the microbiome in IBD pathophysiology. Emerging therapeutic strategies, including probiotics, prebiotics, and dietary interventions, aim to restore microbial balance and mitigate inflammation. Advanced studies on microbiota-targeted therapies reveal their potential to reduce disease severity and improve patient outcomes. Nevertheless, further research is needed to elucidate the bidirectional interactions between the gut microbiome and host immune responses and to translate these insights into clinical applications. This review consolidates current findings on the gut microbiota's role in IBD, emphasizing its diagnostic and therapeutic implications, and advocates for the continued exploration of microbiome-based interventions to combat this debilitating disease.
Collapse
Affiliation(s)
- Bharti Sharma
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - George Agriantonis
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Kate Twelker
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Danielle Ebelle
- Department of Medicine, St. George’s University, Grenada FZ818, West Indies; (D.E.); (M.S.); (W.S.); (J.C.)
| | - Samantha Kiernan
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
| | - Maham Siddiqui
- Department of Medicine, St. George’s University, Grenada FZ818, West Indies; (D.E.); (M.S.); (W.S.); (J.C.)
| | - Aditi Soni
- Department of Medicine, St. George’s University, Grenada FZ818, West Indies; (D.E.); (M.S.); (W.S.); (J.C.)
| | - Sittha Cheerasarn
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
| | - Whenzdjyny Simon
- Department of Medicine, St. George’s University, Grenada FZ818, West Indies; (D.E.); (M.S.); (W.S.); (J.C.)
| | - Winston Jiang
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Angie Cardona
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
| | - Jessica Chapelet
- Department of Medicine, St. George’s University, Grenada FZ818, West Indies; (D.E.); (M.S.); (W.S.); (J.C.)
| | - Alexandra Z. Agathis
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Alejandro Gamboa
- Department of Medicine, Medical University of the Americas, Devens, MA 01434, USA;
| | - Jasmine Dave
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Juan Mestre
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Navin D. Bhatia
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Zahra Shaefee
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Jennifer Whittington
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| |
Collapse
|
77
|
Hou J, Du Y, Cui H. RNF6 promotes cell proliferation of glioblastoma by targeting ubiquitin-mediated degradation of p27. Biochem Biophys Res Commun 2025; 752:151460. [PMID: 39938447 DOI: 10.1016/j.bbrc.2025.151460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
RNF6 (RING finger protein 6), an atypical RING-type ubiquitin ligase, has been reported to be a potential tumor promoter in several human cancers. However, the role of RNF6 in glioblastoma remains poorly understood. In this study, we found that RNF6 was highly expressed in glioblastoma tissues, and its elevated expression was significantly associated with poor prognosis in glioblastoma patients. RNF6 depletion remarkably inhibited cell growth of glioblastoma cells. Mechanistically, RNF6 depletion stabilized p27 protein expression. We demonstrated that RNF6 interacted with p27 and mediated its ubiquitination and degradation in an E3 ligase activity-dependent manner. Moreover, we provide the first evidence revealing the crucial role RNF6 in mediating SKP2 expression at both transcriptional and post-translational levels. On the one hand, RNF6 played as a transcription factor to regulate the activity of the SKP2 promoter. On the other hand, RNF6 interacted with SKP2 and stabilized its protein levels in an Akt-dependent manner. Collectively, our data indicated that RNF6 accelerated glioblastoma cell proliferation and tumorigenesis by targeting p27 for degradation.
Collapse
Affiliation(s)
- Jianbing Hou
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China; Jinfeng Laboratory, Chongqing, 400716, China
| | - Yi Du
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China; Jinfeng Laboratory, Chongqing, 400716, China.
| |
Collapse
|
78
|
Welfer GA, Brady RA, Natchiar SK, Watson ZL, Rundlet EJ, Alejo JL, Singh AP, Mishra NK, Altman RB, Blanchard SC. Impacts of ribosomal RNA sequence variation on gene expression and phenotype. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230379. [PMID: 40045785 PMCID: PMC11883441 DOI: 10.1098/rstb.2023.0379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 11/19/2024] [Accepted: 01/06/2025] [Indexed: 03/09/2025] Open
Abstract
Since the framing of the Central Dogma, it has been speculated that physically distinct ribosomes within cells may influence gene expression and cellular physiology. While heterogeneity in ribosome composition has been reported in bacteria, protozoans, fungi, zebrafish, mice and humans, its functional implications remain actively debated. Here, we review recent evidence demonstrating that expression of conserved variant ribosomal DNA (rDNA) alleles in bacteria, mice and humans renders their actively translating ribosome pool intrinsically heterogeneous at the level of ribosomal RNA (rRNA). In this context, we discuss reports that nutrient limitation-induced stress in Escherichia coli leads to changes in variant rRNA allele expression, programmatically altering transcription and cellular phenotype. We highlight that cells expressing ribosomes from distinct operons exhibit distinct drug sensitivities, which can be recapitulated in vitro and potentially rationalized by subtle perturbations in ribosome structure or in their dynamic properties. Finally, we discuss evidence that differential expression of variant rDNA alleles results in different populations of ribosome subtypes within mammalian tissues. These findings motivate further research into the impacts of rRNA heterogeneities on ribosomal function and predict that strategies targeting distinct ribosome subtypes may hold therapeutic potential.This article is part of the discussion meeting issue 'Ribosome diversity and its impact on protein synthesis, development and disease'.
Collapse
Affiliation(s)
- Griffin A. Welfer
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN38105, USA
| | - Ryan A. Brady
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN38105, USA
| | - S. Kundhavai Natchiar
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN38105, USA
| | - Zoe L. Watson
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN38105, USA
| | - Emily J. Rundlet
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712, USA
| | - Jose L. Alejo
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN38105, USA
| | - Anand P. Singh
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN38105, USA
| | - Nitish K. Mishra
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN38105, USA
| | - Roger B. Altman
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN38105, USA
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105, USA
| | - Scott C. Blanchard
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN38105, USA
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105, USA
| |
Collapse
|
79
|
Zou B, Liu S, Dong C, Shen H, Lv Y, He J, Li X, Ruan M, Huang Z, Shu S. Fecal microbiota transplantation restores gut microbiota diversity in children with active Crohn's disease: a prospective trial. J Transl Med 2025; 23:288. [PMID: 40050917 PMCID: PMC11887145 DOI: 10.1186/s12967-024-05832-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 10/31/2024] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Clinical data on oral fecal microbiota transplantation (FMT), a promising therapy for Crohn's disease (CD), are limited. Herein, we determined the short-term safety and feasibility of FMT for pediatric patients with active CD. METHODS In this open-label, parallel-group, single-center prospective trial, patients with active CD were treated with oral FMT capsules combined with partial enteral nutrition (PEN) (80%). The control group comprised pediatric patients with active CD treated with PEN (80%) and immunosuppressants. Thirty-three patients (11.6 ± 1.82 years)-17 in the capsule and 16 in the control groups-were analyzed. Data regarding the adverse events, clinical reactions, intestinal microbiome composition, and biomarker parameters were collected and compared post-treatment. RESULTS At week 10, the clinical and endoscopic remission rates did not differ between the two groups. By week 10, the mean fecal calprotectin level, C-reactive protein level, erythrocyte sedimentation rate, simple endoscopic score for CD, and pediatric CD activity index decreased significantly in the capsule group (all P < 0.05). The main adverse event was mild-to-moderate constipation. Core functional genera, Agathobacter, Akkermansia, Roseburia, Blautia, Subdoligranulum, and Faecalibacterium, were lacking pre-treatment. Post-treatment, the implantation rates of these core functional genera increased significantly, which positively correlated with the anti-inflammatory factor, interleukin (IL)-10, and negatively correlated with the pro-inflammatory factor, IL-6. The combination of these six functional genera distinguished healthy children from those with CD (area under the curve = 0.96). CONCLUSIONS Oral FMT capsules combined with PEN (80%) could be an effective therapy for children with active CD. The six core functional genera identified here may be candidate biomarkers for identifying children with CD. TRIAL REGISTRATION ClinicalTrials.gov, retrospectively registered, ID# NCT05321758, NCT05321745, date of registration: 2022-04-04.
Collapse
Affiliation(s)
- Biao Zou
- Pediatric Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan, 430030, Hubei, China
| | - Shengxuan Liu
- Pediatric Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan, 430030, Hubei, China
| | - Chen Dong
- Pediatric Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan, 430030, Hubei, China
| | - Hexiao Shen
- School of Life Science, Hubei University, Wuhan, 430030, Hubei, China
| | - Yongling Lv
- School of Life Science, Hubei University, Wuhan, 430030, Hubei, China
| | - Jiayi He
- Pediatric Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan, 430030, Hubei, China
| | - Xuesong Li
- Pediatric Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan, 430030, Hubei, China
| | - Mengling Ruan
- Pediatric Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan, 430030, Hubei, China
| | - Zhihua Huang
- Pediatric Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan, 430030, Hubei, China
| | - Sainan Shu
- Pediatric Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan, 430030, Hubei, China.
| |
Collapse
|
80
|
Cremer A, Rosewick N, Kelsey M, Trépo E, Libert F, De Vos M, Baert F, Moreels T, Louis E, Rahier JF, Demetter P, Sedivy JM, Vermeire S, Franchimont D. A transcriptomic score to classify the inflammation-dysplasia-cancer sequence lesions in inflammatory bowel disease. J Crohns Colitis 2025; 19:jjaf026. [PMID: 39945142 PMCID: PMC11904305 DOI: 10.1093/ecco-jcc/jjaf026] [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] [Indexed: 03/14/2025]
Abstract
BACKGROUND AND AIMS Inflammatory bowel disease (IBD) is associated with a higher risk of developing colorectal cancer, according to the inflammation-dysplasia-cancer (IDC) sequence from inflammation to colitis-associated colorectal cancer (CAC). The objective of this study was to identify and generate a transcriptomic signature and score, related to the IDC sequence, that could ultimately classify dysplasia and cancer in IBD. METHODS Demographics, clinical parameters, histological characteristics, and RNA-sequencing data were evaluated on 134 formalin-fixed paraffin-embedded lesions from 2 independent cohorts of IBD patients with low- or high-grade dysplasia (LGD, HGD) and/or CAC. An ordinal logistic regression screened for significant IDC sequence-associated genes that were computed in a transcriptomic signature score. RESULTS Principal component analysis and unsupervised clustering on 1% of the most variable genes showed a good clustering between the 4 lesion groups (Normal Mucosa, Inflamed Mucosa, LGD/HGD, and CAC). A gene signature was identified on 27 genes that correlated with the lesion groups in the exploratory cohort. The most weighted gene in this transcriptomic signature was the long non-coding regulatory RNA KCNQ1OT1, a gatekeeper against genomic instability and transposon activation. Based on the expression of these 27 genes, we built and validated a transcriptomic signature score to classify dysplasia and CAC. The overall accuracy of the transcriptomic signature score was 85.71% in the exploratory cohort and 90.91% in the validation cohort. CONCLUSION We identified a tissue-based transcriptomic score to classify IDC lesions in IBD patients and uncovered some of the pivotal genes in carcinogenesis related to inflammation in IBD.
Collapse
Affiliation(s)
- Anneline Cremer
- Department of Gastroenterology, HUB Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicolas Rosewick
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Maxfield Kelsey
- Center on the Biology of Aging, and the Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Eric Trépo
- Department of Gastroenterology, HUB Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Frédérick Libert
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Martine De Vos
- Department of Gastroenterology, University Hospital Ghent, Ghent, Belgium
| | - Filip Baert
- Department of Gastroenterology, AZ Delta, Roeselare, Belgium
| | - Tom Moreels
- Department of Gastroenterology, University Hospital Antwerp, Edegem, Belgium
| | - Edouard Louis
- Department of Gastroenterology, University Hospital Liège, Liège, Belgium
| | - Jean-François Rahier
- Department of Gastroenterology, CHU UCL Namur site Mont-Godinne, Université Catholique de Louvain, Yvoir, Belgium
| | - Pieter Demetter
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - John M Sedivy
- Center on the Biology of Aging, and the Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Séverine Vermeire
- Department of Gastroenterology, University Hospital Leuven, Leuven, Belgium
| | - Denis Franchimont
- Department of Gastroenterology, HUB Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
81
|
Krynicka P, Kaczmarczyk M, Skonieczna-Żydecka K, Styburski D, Podsiadło K, Cembrowska-Lech D, Dąbkowski K, Deskur A, Rogoza-Mateja W, Ławniczak M, Białek A, Koulaouzidis A, Marlicz W. Untargeted Metabolomic Profiling of Colonic Mucosa in Individuals with Irritable Bowel Syndrome. Biomedicines 2025; 13:629. [PMID: 40149605 PMCID: PMC11940239 DOI: 10.3390/biomedicines13030629] [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: 02/05/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Irritable Bowel Syndrome (IBS) is a complex disorder characterized by altered gut-brain interactions, with gastrointestinal microbiota and metabolic dysregulation playing key roles in its pathophysiology. Identifying specific metabolic alterations within the colonic mucosa may enhance our understanding of IBS and contribute to improved diagnostic and therapeutic approaches. Methods: This cross-sectional study analyzed the metabolomic profiles of colonic mucosal biopsies from 44 IBS patients assessed with ROME IV criteria and 69 healthy controls undergoing colonoscopy. Untargeted metabolomic profiling was conducted using liquid chromatography-mass spectrometry (LC-MS), and differential metabolite analysis was performed via fold-change calculations and machine learning-based classification. Results: IBS patients exhibited distinct mucosal metabolic profiles, with significantly elevated levels of N-acetylneuraminic acid and 1-palmitoylglycerol, suggesting compromised epithelial integrity and increased gut permeability. In contrast, cis-4-hydroxycyclohexanecarboxylic acid, a metabolite associated with protective mucosal functions, was reduced. Random Forest analysis identified these metabolites as key discriminatory features between IBS and control groups, reinforcing their potential role as biomarkers for IBS-related mucosal alterations. Conclusions: Our study highlights the unique metabolomic signatures of IBS at the mucosal level, emphasizing the role of microbial metabolites in disease pathology. These findings may facilitate the development of novel diagnostic tools and targeted therapeutic strategies, advancing personalized management for IBS patients.
Collapse
Affiliation(s)
- Patrycja Krynicka
- Department of Gastroenterology, Pomeranian Medical University, 71-252 Szczecin, Poland; (P.K.); (W.R.-M.); (A.B.); (W.M.)
| | - Mariusz Kaczmarczyk
- Department of Biochemical Science, Pomeranian Medical University, 71-460 Szczecin, Poland (K.S.-Ż.)
- Sanprobi sp. z o.o. sp.k., 70-525 Szczecin, Poland (K.P.); (D.C.-L.)
| | - Karolina Skonieczna-Żydecka
- Department of Biochemical Science, Pomeranian Medical University, 71-460 Szczecin, Poland (K.S.-Ż.)
- Sanprobi sp. z o.o. sp.k., 70-525 Szczecin, Poland (K.P.); (D.C.-L.)
| | - Daniel Styburski
- Sanprobi sp. z o.o. sp.k., 70-525 Szczecin, Poland (K.P.); (D.C.-L.)
| | - Konrad Podsiadło
- Sanprobi sp. z o.o. sp.k., 70-525 Szczecin, Poland (K.P.); (D.C.-L.)
| | | | - Krzysztof Dąbkowski
- Department of Gastroenterology, Pomeranian Medical University, 71-252 Szczecin, Poland; (P.K.); (W.R.-M.); (A.B.); (W.M.)
| | - Anna Deskur
- Department of Gastroenterology, Pomeranian Medical University, 71-252 Szczecin, Poland; (P.K.); (W.R.-M.); (A.B.); (W.M.)
| | - Wiesława Rogoza-Mateja
- Department of Gastroenterology, Pomeranian Medical University, 71-252 Szczecin, Poland; (P.K.); (W.R.-M.); (A.B.); (W.M.)
| | - Małgorzata Ławniczak
- Department of Gastroenterology, Pomeranian Medical University, 71-252 Szczecin, Poland; (P.K.); (W.R.-M.); (A.B.); (W.M.)
| | - Andrzej Białek
- Department of Gastroenterology, Pomeranian Medical University, 71-252 Szczecin, Poland; (P.K.); (W.R.-M.); (A.B.); (W.M.)
| | - Anastasios Koulaouzidis
- Department of Gastroenterology, Pomeranian Medical University, 71-252 Szczecin, Poland; (P.K.); (W.R.-M.); (A.B.); (W.M.)
- Department of Clinical Research, University of Southern Denmark (SDU), 5230 Odense, Denmark
- Research Unit, Department of Surgery, Odense University Hospital (OUH), 5000 Svendborg, Denmark
| | - Wojciech Marlicz
- Department of Gastroenterology, Pomeranian Medical University, 71-252 Szczecin, Poland; (P.K.); (W.R.-M.); (A.B.); (W.M.)
- Sanprobi sp. z o.o. sp.k., 70-525 Szczecin, Poland (K.P.); (D.C.-L.)
| |
Collapse
|
82
|
Carneiro KDO, Araújo TMT, Da Silva Mourão RM, Casseb SMM, Demachki S, Moreira FC, Dos Santos ÂKCR, Ishak G, Da Costa DDSA, Magalhães L, Vidal AF, Burbano RMR, de Assumpção PP. Transcriptional and microbial profile of gastric cancer patients infected with Epstein-Barr virus. Front Oncol 2025; 15:1530430. [PMID: 40110195 PMCID: PMC11919665 DOI: 10.3389/fonc.2025.1530430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Gastric cancer (GC), which has low survival rates and high mortality, is a major concern, particularly in Asia and South America, with over one million annual cases. Epstein-Barr virus (EBV) is recognized as a carcinogen that may trigger gastric carcinogenesis by infecting the stomach epithelium via reactivated B cells, with growing evidence linking it to GC. This study investigates the transcriptional and microbial profiles of EBV-infected versus EBV-non-infected GC patients. Methods Using Illumina NextSeq, cDNA libraries were sequenced, and reads were aligned to the human genome and analyzed with DESeq2. Kegg and differential analyses revealed key genes and pathways. Gene sensitivity and specificity were assessed using ROC curves (p < 0.05, AUC > 0.8). Non-aligned reads were used for microbiome analysis with Kraken2 for bacterial identification. Microbial analysis included LDA score, Alpha and Beta diversity metrics, with significance set at p ≤ 0.05. Spearman's correlation between differentially expressed genes (DEGs) and bacteria were also examined. Results The data revealed a gene expression pattern in EBV-positive gastric cancer, highlighting immune response, inflammation, and cell proliferation genes (e.g., GBP4, ICAM1, IL32, TNFSF10). ROC analysis identified genes with high specificity and sensitivity for discriminating EBV+ gastric cancer, including GBP5, CMKLR1, GM2A and CXCL11 that play pivotal roles in immune response, inflammation, and cancer. Functional enrichment pointed to cytokine-cytokine receptor interactions, antigen processing, and Th17 immune response, emphasizing the role of the tumor microenvironment, shaped by inflammation and immunomodulation, in EBV-associated GC. Microbial analysis revealed changes in the gastric microbiota in EBV+ samples, with a significant reduction in bacterial taxa. The genera Choristoneura and Bartonella were more abundant in EBV+ GC, while more abundant bacteria in EBV- GC included Citrobacter, Acidithiobacillus and Biochmannia. Spearman's correlation showed a strong link between DE bacterial genera and DEGs involved in processes like cell differentiation, cytokine production, digestion, and cell death. Conclusion These findings suggest a complex interaction between the host (EBV+ GC) and the microbiota, possibly influencing cancer progression, and offering potential therapeutic targets such as microbiota modulation or gene regulation. Comparing with EBV- samples further highlights the specific impact of EBV and the microbiota on gastric cancer pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Samia Demachki
- Oncology Research Center, Federal University of Pará, Belém, Brazil
| | | | | | - Geraldo Ishak
- Oncology Research Center, Federal University of Pará, Belém, Brazil
| | | | | | | | | | | |
Collapse
|
83
|
Bakir-Gungor B, Temiz M, Canakcimaksutoglu B, Yousef M. Prediction of colorectal cancer based on taxonomic levels of microorganisms and discovery of taxonomic biomarkers using the Grouping-Scoring-Modeling (G-S-M) approach. Comput Biol Med 2025; 187:109813. [PMID: 39929003 DOI: 10.1016/j.compbiomed.2025.109813] [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: 06/12/2024] [Revised: 01/09/2025] [Accepted: 02/05/2025] [Indexed: 02/12/2025]
Abstract
Colorectal cancer (CRC) is one of the most prevalent forms of cancer globally. The human gut microbiome plays an important role in the development of CRC and serves as a biomarker for early detection and treatment. This research effort focuses on the identification of potential taxonomic biomarkers of CRC using a grouping-based feature selection method. Additionally, this study investigates the effect of incorporating biological domain knowledge into the feature selection process while identifying CRC-associated microorganisms. Conventional feature selection techniques often fail to leverage existing biological knowledge during metagenomic data analysis. To address this gap, we propose taxonomy-based Grouping Scoring Modeling (G-S-M) method that integrates biological domain knowledge into feature grouping and selection. In this study, using metagenomic data related to CRC, classification is performed at three taxonomic levels (genus, family and order). The MetaPhlAn tool is employed to determine the relative abundance values of species in each sample. Comparative performance analyses involve six feature selection methods and four classification algorithms. When experimented on two CRC associated metagenomics datasets, the highest performance metric, yielding an AUC of 0.90, is observed at the genus taxonomic level. At this level, 7 out of top 10 groups (Parvimonas, Peptostreptococcus, Fusobacterium, Gemella, Streptococcus, Porphyromonas and Solobacterium) were commonly identified for both datasets. Moreover, the identified microorganisms at genus, family, and order levels are thoroughly discussed via refering to CRC-related metagenomic literature. This study not only contributes to our understanding of CRC development, but also highlights the applicability of taxonomy-based G-S-M method in tackling various diseases.
Collapse
Affiliation(s)
- Burcu Bakir-Gungor
- Department of Computer Engineering, Faculty of Engineering, Abdullah Gul University, Kayseri, 38080, Turkey
| | - Mustafa Temiz
- Department of Electrical and Computer Engineering, Faculty of Engineering, Abdullah Gul University, Kayseri, 38080, Turkey.
| | - Beyza Canakcimaksutoglu
- Department of Bioengineering, Faculty of Life and Natural Science, Abdullah Gul University, Kayseri, 38080, Turkey
| | - Malik Yousef
- Department of Information Systems, Zefat Academic College, Zefat, 13206, Israel; Galilee Digital Health Research Center (GDH), Zefat Academic College, Israel
| |
Collapse
|
84
|
Zhao B, Zhang B, Chenzhang M, Jiang K, Wang D, Chen J. Ethacrynic acid inhibits the growth and proliferation of prostate cancer cells by targeting GSTP1 and regulating the PI3K-AKT signaling pathway. Int J Biochem Cell Biol 2025; 180:106740. [PMID: 39900237 DOI: 10.1016/j.biocel.2025.106740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 01/09/2025] [Accepted: 01/22/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND As a diuretic, ethacrynic acid (EA) has been shown to play a suppressive role in cancers, including prostate cancer (PC). However, its molecular regulatory mechanism is still unclear. Therefore, our study is centered on investigating the effect of EA on PC development and its mechanism. METHODS To verify the binding relationship between EA and GSTP1, molecular docking and cellular thermal shift assay (CETSA) were conducted. To examine how EA affects PC cell proliferation, cell cycle, and apoptosis, cell function assays were performed. qRT-PCR was used to detect GSTP1 mRNA expression. The expression of GSTP1 protein and PI3K-AKT signaling pathway-related proteins in cells was detected by western blot (WB). To verify how EA and GSTP1 influence cell growth in PC, in vivo experiments were conducted. RESULTS The binding relationship between GSTP1 and EA was confirmed by molecular docking and CETSA results. Cell experiments showed that EA could hinder PI3K/AKT pathway and PC cell proliferation, arrest the cell cycle in G0/G1 phase, and facilitate apoptosis by binding to GSTP1. In vivo experiments in nude mice verified that the interaction between EA and GSTP1 reduced PI3K and AKT phosphorylation and inhibited the growth of PC cells. CONCLUSION EA inhibits PC progression by binding to GSTP1 to downregulate the activity of PI3K/AKT pathway, and this result suggests the potential of EA to be an anticancer agent for PC therapy.
Collapse
Affiliation(s)
- Bin Zhao
- Department of Urology, People's Hospital Affiliated of Quanzhou Medical College, Quanzhou 362000, China; The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000, China
| | - Bingfeng Zhang
- The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000, China
| | - Minhao Chenzhang
- The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000, China
| | - Kangxian Jiang
- The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000, China
| | - Dianyu Wang
- The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000, China
| | - Junyi Chen
- The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000, China; Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China.
| |
Collapse
|
85
|
Guo X, Du X, Zhao G, Liu C, Gao J, Huang Z, Dong W. OSR1 suppresses oral squamous cell carcinoma proliferation and migration via the AXIN2/β-catenin pathway. Oral Dis 2025; 31:741-755. [PMID: 39286942 DOI: 10.1111/odi.15135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/10/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVES The odd-skipped related transcription factor 1 (OSR1) gene exerts distinct regulatory effects on tumorigenesis and development in various cancer types. However, the precise role of OSR1 in oral squamous cell carcinoma (OSCC) remains to be elucidated. METHODS GEPIA 2 and TCGA databases were utilized to analyze the OSR1 expression in head and neck squamous cell carcinoma (HNSC) patients and its impact on prognosis. Hematoxylin-eosin staining, immunohistochemistry, immunofluorescence, western blotting, and RT-qPCR were employed to detect the OSR1 expression in OSCC tissues and cells. Lentivirus transfection was utilized for overexpression and downexpression of OSR1 in OSCC. CCK8 cell proliferation assay, colony formation and cell scratch assay were conducted to investigate the effects of OSR1 on biological behavior of OSCC cells. Western blotting and RT-qPCR were applied to investigate the regulatory mechanism of OSR1 on AXIN2/β-catenin signaling pathway. RESULTS OSR1 expression was significantly decreased in HNSC patients, OSCC tissues and cells, leading to a decrease in 5-year survival rate. OSR1 overexpression inhibited the proliferation and migration of OSCC cells, and the AXIN2/β-catenin signaling pathway was inhibited. Silencing OSR1 had the opposite effect. CONCLUSIONS OSR1 functioned as a tumor suppressor gene in OSCC proliferation and migration by regulating the AXIN2/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xintong Guo
- School of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Xinyi Du
- School of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Gaoye Zhao
- School of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Chongshen Liu
- School of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Jing Gao
- School of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Zunzhi Huang
- School of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Wei Dong
- School of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| |
Collapse
|
86
|
Jiang M, Zhang R, Huang M, Yang J, Liu Q, Zhao Z, Ma Y, Zhao H, Zhang M. The Prognostic Value of Tumor-Associated Neutrophils in Colorectal Cancer: A Systematic Review and Meta-Analysis. Cancer Med 2025; 14:e70614. [PMID: 40013340 PMCID: PMC11865885 DOI: 10.1002/cam4.70614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 06/23/2024] [Accepted: 01/06/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Tumor-associated neutrophils (TANs) are important components of the colorectal cancer (CRC) microenvironment. However, their role in CRC remains controversial. This study aimed to assess the prognostic value of TANs in patients with CRC. METHODS We searched the PubMed, EMBASE, and Cochrane Library databases for eligible studies published until January 9, 2023. The pooled hazard ratios (HRs) and odds ratios (ORs) with their 95% confidence intervals (95% CI) were calculated with a random-effects model to assess survival outcomes and clinicopathological features. Subgroup analyses were further conducted to identify potential sources of heterogeneity. Funnel plots and Egger's test were used to measure publication bias. RESULTS A total of 19 studies with 7721 patients were included in this meta-analysis. The pooled analysis indicated that high peritumoral TAN infiltration in CRC tissue was significantly associated with favorable cancer-specific survival (HR = 0.57; 95% CI: 0.38-0.86; p = 0.007), but not with overall survival or disease-free survival. No association between high intratumoral or unclear compartment TAN infiltration and CRC prognosis was found. Subgroup analyses showed that the association between TANs and the prognosis of CRC patients differed according to antibody types, tumor stage, quantitative methods, and follow-up time. High intratumoral TAN infiltration was significantly associated with histology type, whereas high TAN infiltration in an unclear compartment was significantly associated with gender, tumor location, and the primary tumor site. CONCLUSIONS High TAN infiltration, especially in the peritumoral compartment, could be a potential prognostic marker in CRC. More high-quality studies are required to explore its specific prognostic value in CRC.
Collapse
Affiliation(s)
- Mengyuan Jiang
- Department of PathologyTaicang Loujiang New City HospitalTaicangJiangsuChina
- Department of PathologyGansu Provincial HospitalLanzhouGansuChina
- The First School of Clinical MedicineGansu University of Traditional Chinese MedicineLanzhouGansuChina
- Department of PathologyThe 940th Hospital of Joint Logistics Support Force of Chinese People´s Liberation ArmyLanzhouGansuChina
| | - Rui Zhang
- The First School of Clinical MedicineGansu University of Traditional Chinese MedicineLanzhouGansuChina
- Department of PathologyThe 940th Hospital of Joint Logistics Support Force of Chinese People´s Liberation ArmyLanzhouGansuChina
- Department of PathologyGansu Provincial Cancer HospitalLanzhouGansuChina
| | - Min Huang
- The First School of Clinical MedicineGansu University of Traditional Chinese MedicineLanzhouGansuChina
- Department of PathologyThe 940th Hospital of Joint Logistics Support Force of Chinese People´s Liberation ArmyLanzhouGansuChina
| | - Jing Yang
- The First School of Clinical MedicineGansu University of Traditional Chinese MedicineLanzhouGansuChina
- Department of PathologyChengdu First People's HospitalChengduSichuanChina
| | - Qianqian Liu
- The First School of Clinical MedicineGansu University of Traditional Chinese MedicineLanzhouGansuChina
| | - Ziru Zhao
- The First School of Clinical MedicineGansu University of Traditional Chinese MedicineLanzhouGansuChina
| | - Ya Ma
- Department of PathologyGansu Provincial HospitalLanzhouGansuChina
- The First School of Clinical MedicineGansu University of Traditional Chinese MedicineLanzhouGansuChina
| | - Hongfan Zhao
- Department of PathologyGansu Provincial HospitalLanzhouGansuChina
- The First School of Clinical MedicineLanzhou UniversityLanzhouGansuChina
| | - Min Zhang
- Department of PathologyGansu Provincial HospitalLanzhouGansuChina
- The First School of Clinical MedicineGansu University of Traditional Chinese MedicineLanzhouGansuChina
- Clinical Research Centre, Department of Science and TechnologySichuan Provincial People's Hospital, University of Electronic Science and Technology of ChinaChengduSichuanChina
| |
Collapse
|
87
|
Bosco V, Mercuri C, Doldo P, Nocerino R, Guillari A, Virgolesi M, Rea T, Giordano V, Simeone S. The Lived Experience of Adults With Inflammatory Bowel Disease in Rural Areas: Phenomenological Study. Nurs Health Sci 2025; 27:e70058. [PMID: 39928987 PMCID: PMC11810439 DOI: 10.1111/nhs.70058] [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: 06/11/2024] [Revised: 01/02/2025] [Accepted: 01/26/2025] [Indexed: 02/12/2025]
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a chronic gastrointestinal condition with a multifactorial etiology. It significantly impacts patients' quality of life, particularly, in rural areas where access to specialized care is limited. Challenges such as increased travel costs, social isolation, and restricted healthcare access are recognized, but their specific impact on rural Italian patients remains underexplored. This study aims to fill this gap. Using Cohen's hermeneutic phenomenological approach, in-depth interviews were conducted with 17 IBD patients from rural areas. Thematic analysis identified key patterns and themes. Two main themes emerged: "challenges of rural living," including economic concerns and social isolation, and "communication that bridges distances." Rural IBD patients face unique challenges extending beyond disease management, with economic hardship and social isolation being prominent. These findings highlight the need for tailored interventions to bridge healthcare gaps and improve the quality of life for rural patients, offering novel insights into their lived experiences in Italy.
Collapse
Affiliation(s)
- Vincenzo Bosco
- Department of Medical and Surgical Sciences, University Hospital Mater DominiMagna Graecia UniversityCatanzaroItaly
| | - Caterina Mercuri
- Department of Clinical and Experimental MedicineUniversity of Catanzaro Magna GraeciaCatanzaroItaly
| | - Patrizia Doldo
- Department of Clinical and Experimental MedicineUniversity of Catanzaro Magna GraeciaCatanzaroItaly
| | - Rita Nocerino
- Department of Translational Medical ScienceUniversity of Naples “Federico II”NaplesItaly
- ImmunoNutritionLab at CEINGE—Advanced BiotechnologiesUniversity of Naples “Federico II”NaplesItaly
| | - Assunta Guillari
- Department of Public HealthUniversity of Naples “Federico II”NaplesItaly
| | - Michele Virgolesi
- Department of Public HealthUniversity of Naples “Federico II”NaplesItaly
| | - Teresa Rea
- Department of Public HealthUniversity of Naples “Federico II”NaplesItaly
| | - Vincenza Giordano
- Department of General Surgery and Women's HealthAORN Antonio CardarelliNaplesItaly
| | - Silvio Simeone
- Department of Clinical and Experimental MedicineUniversity of Catanzaro Magna GraeciaCatanzaroItaly
| |
Collapse
|
88
|
Wu X, Zhang Y, Ding Y, Yang J, Song Z, Lin S, Zhang R, Wu J, Shen S. Nanosize Non-Viral Gene Therapy Reverses Senescence Reprograming Driven by PBRM1 Deficiency to Suppress iCCA Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414525. [PMID: 39823528 PMCID: PMC11904949 DOI: 10.1002/advs.202414525] [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: 11/07/2024] [Revised: 12/28/2024] [Indexed: 01/19/2025]
Abstract
Polybromo-1 (PBRM1) serves as a crucial regulator of gene transcription in various tumors, including intrahepatic cholangiocarcinoma (iCCA). However, the exact role of PBRM1 in iCCA and the mechanism by which it regulates downstream target genes remain unclear. This research has revealed that PBRM1 is significantly downregulated in iCCA tissues, and this reduced expression is linked to aggressive clinicopathological features and a poor prognosis. Furthermore, it is demonstrated that PBRM1 can impede iCCA progression, and a gene therapy nanomedicine is developed to treat iCCA in vivo by modulating PBRM1 expression. The heightened expression of PBRM1 induces by the nanomedicine substantially inhibited tumor growth in iCCA. Conversely, the decrease in PBRM1 results in the abnormal activation of the ERK1/2 signaling pathway, a reduction in p16, p53/p21, and cellular senescence, thereby promoting iCCA advancement. Treatment with U0126, an ERK1/2 inhibitor, effectively halted iCCA progression by regulating the PBRM1-ERK1/2-cellular senescence pathway. These findings underscore the significant role of PBRM1 in controlling iCCA progression and predicting prognosis. Targeting the PBRM1-ERK1/2-cellular senescence pathway with U0126 shows promise for clinical applications in treating iCCA.
Collapse
Affiliation(s)
- Xiwen Wu
- Department of Hepatic SurgeryCenter of Hepato‐Pancreato‐Biliary SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Department of Clinical NutritionSun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Yi Zhang
- Department of Hepatic SurgeryCenter of Hepato‐Pancreato‐Biliary SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Department of Hepatobiliary SurgeryThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Yuan Ding
- Department of Hepatic SurgeryCenter of Hepato‐Pancreato‐Biliary SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Jiali Yang
- Department of Hepatic SurgeryCenter of Hepato‐Pancreato‐Biliary SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Zimin Song
- Department of Hepatic SurgeryCenter of Hepato‐Pancreato‐Biliary SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Shuirong Lin
- Department of Hepatic SurgeryCenter of Hepato‐Pancreato‐Biliary SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Ruhe Zhang
- Department of HematologyThe Seventh Affiliated Hospital of Sun Yat‐sen UniversityShenzhen518107China
| | - Jun Wu
- Bioscience and Biomedical Engineering ThrustThe Hong Kong University of Science and Technology (Guangzhou)NanshaGuangzhouGuangdong511400China
- Division of Life ScienceThe Hong Kong University of Science and TechnologyHong Kong SAR999077China
| | - Shunli Shen
- Department of Hepatic SurgeryCenter of Hepato‐Pancreato‐Biliary SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
| |
Collapse
|
89
|
Pipatthana M, Phanchana M, Sangphukieo A, Charoensutthivarakul S, Harnvoravongchai P, Chankhamhaengdecha S, Prangthip P, Konpetch P, Sripong C, Wongphayak S, Janvilisri T. Repurposing thioridazine as a potential CD2068 inhibitor to mitigate antibiotic resistance in Clostridioides difficile infection. Comput Struct Biotechnol J 2025; 27:887-895. [PMID: 40123799 PMCID: PMC11928863 DOI: 10.1016/j.csbj.2025.02.036] [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: 11/08/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Clostridioides difficile infection (CDI) is a major public health issue, driven by antibiotic resistance and frequent recurrence. CD2068, an ABC protein in C. difficile, is associated with drug resistance, making it a potential target for novel therapies. This study explored FDA-approved non-antibiotic drugs for their ability to inhibit CD2068 through drug screening and experimental validation. Thioridazine exhibited moderate binding affinity to CD2068 and inhibited its ATP hydrolysis activity. It also suppressed the growth of multiple C. difficile ribotypes at 64-128 µg/mL, with rapid-killing effects. When combined with sub-MIC levels of standard antibiotics, thioridazine significantly reduced bacterial growth. In a mouse CDI model, thioridazine demonstrated potential in restoring gut microbial balance and improving survival, although it did not show superiority to vancomycin. These findings suggest that thioridazine has potential as a novel therapeutic for CDI, either as an adjunct to existing antibiotics or as part of a combination therapy to combat antibiotic resistance. Further research, including replication studies and dose optimization, is needed to fully evaluate thioridazine's therapeutic potential.
Collapse
Affiliation(s)
- Methinee Pipatthana
- Department of Microbiology, Faculty of Public Health, Mahidol University, Bangkok, Thailand
| | - Matthew Phanchana
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Apiwat Sangphukieo
- Center of Multidisciplinary Technology for Advanced Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sitthivut Charoensutthivarakul
- Excellence Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, Thailand
- School of Bioinnovation and Bio-Based Product Intelligence, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | | | - Pattaneeya Prangthip
- Department of Tropical Nutrition and Food Science, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Pattanai Konpetch
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | | | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
90
|
Yu T, Chae M, Wang Z, Ryu G, Kim GB, Lee SY. Microbial Technologies Enhanced by Artificial Intelligence for Healthcare Applications. Microb Biotechnol 2025; 18:e70131. [PMID: 40100535 PMCID: PMC11917392 DOI: 10.1111/1751-7915.70131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
The combination of artificial intelligence (AI) with microbial technology marks the start of a major transformation, improving applications throughout biotechnology, especially in healthcare. With the capability of AI to process vast amounts of biological big data, advanced microbial technology allows for a comprehensive understanding of complex biological systems, advancing disease diagnosis, treatment and the development of microbial therapeutics. This mini review explores the impact of AI-integrated microbial technologies in healthcare, highlighting advancements in microbial biomarker-based diagnosis, the development of microbial therapeutics and the microbial production of therapeutic compounds. This exploration promises significant improvements in the design and implementation of health-related solutions, steering a new era in biotechnological applications.
Collapse
Affiliation(s)
- Taeho Yu
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four)KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST)DaejeonRepublic of Korea
- Systems Metabolic Engineering and Systems Healthcare Cross‐Generation Collaborative LaboratoryKAISTDaejeonRepublic of Korea
| | - Minjee Chae
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four)KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST)DaejeonRepublic of Korea
- Systems Metabolic Engineering and Systems Healthcare Cross‐Generation Collaborative LaboratoryKAISTDaejeonRepublic of Korea
- Graduate School of Engineering BiologyKAISTDaejeonRepublic of Korea
| | - Ziling Wang
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four)KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST)DaejeonRepublic of Korea
- Systems Metabolic Engineering and Systems Healthcare Cross‐Generation Collaborative LaboratoryKAISTDaejeonRepublic of Korea
| | - Gahyeon Ryu
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four)KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST)DaejeonRepublic of Korea
- Systems Metabolic Engineering and Systems Healthcare Cross‐Generation Collaborative LaboratoryKAISTDaejeonRepublic of Korea
| | - Gi Bae Kim
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four)KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST)DaejeonRepublic of Korea
- Systems Metabolic Engineering and Systems Healthcare Cross‐Generation Collaborative LaboratoryKAISTDaejeonRepublic of Korea
- BioProcess Engineering Research CenterKAISTDaejeonRepublic of Korea
| | - Sang Yup Lee
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four)KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST)DaejeonRepublic of Korea
- Systems Metabolic Engineering and Systems Healthcare Cross‐Generation Collaborative LaboratoryKAISTDaejeonRepublic of Korea
- Graduate School of Engineering BiologyKAISTDaejeonRepublic of Korea
- BioProcess Engineering Research CenterKAISTDaejeonRepublic of Korea
- Center for Synthetic BiologyKAISTDaejeonRepublic of Korea
| |
Collapse
|
91
|
Urrutia-Pereira M, Fogelbach GG, Chong-Neto HJ, Solé D. Food additives and their impact on human health. Allergol Immunopathol (Madr) 2025; 53:26-31. [PMID: 40088018 DOI: 10.15586/aei.v53i2.1149] [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: 06/08/2024] [Accepted: 09/15/2024] [Indexed: 03/17/2025]
Abstract
Increasing evidence suggests that high consumption of ultra-processed foods (UPF) is associated with an increase in noncommunicable diseases, overweight, and obesity. This review aimed to verify the association of UPF with inflammatory diseases, especially allergic diseases. To identify relevant articles, an extensive literature search was conducted using the two most important search sites - PubMed and Google Scholar. Specific Medical Subject Headings (MeSHes) such as "food additives and health," "food additives and immune system," and "food additives and diseases" were used to conduct an advanced search. Emulsifiers have been, particularly, implicated in disrupting intestinal barrier function, modifying gut microbiota, and promoting inflammation, which may contribute to the development of food allergies and inflammatory diseases. While food additives serve various functions in the food industry, concerns regarding their impact on health, particularly in systemic autoimmune and metabolic conditions, have been raised. Common additives have been associated with allergic reactions, intolerances, and sensitivities.
Collapse
Affiliation(s)
- Marilyn Urrutia-Pereira
- Department of Pediatrics, Faculty of Medicine, Federal University of Pampa, Uruguaiana RS, Brazil
| | | | - Herberto José Chong-Neto
- Department of Pediatrics, Division of Allergy and Immunology, Federal University of Paraná, Curitiba, Brazil;
| | - Dirceu Solé
- Department of Pediatrics, Division of Allergy, Clinical Immunology and Rheumatology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
92
|
Xiong N, Zhang W, Zhang Y, Nie C, Dan H. Association between nutrient intake and inflammatory bowel disease risk: Insights from NHANES data and dose-response analysis. Nutrition 2025; 131:112632. [PMID: 39700661 DOI: 10.1016/j.nut.2024.112632] [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: 06/27/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND The role of dietary and nutritional factors in inflammatory bowel disease (IBD) remains poorly understood, necessitating further investigation. This study aims to explore the association between nutrient intake and the risk of IBD. METHODS This cross-sectional study utilized data from the 2009-2010 NHANES cycle, focusing on individuals with complete 24-hour dietary intake records and clinically assessed IBD information. Nutrient intake was assessed through dietary recalls and supplement data. Associations between nutrient intake and IBD risk were analyzed by propensity score matching (PSM) with balanced baseline characteristics and logistic regression. Dose-response relationships were examined by restricted cubic splines (RCS). Statistical significance was set at P < 0.05, and analyses were conducted using R software. RESULTS The study included 4,072 participants with clinically assessed IBD and complete nutrient intake data. In adjusted analyses, lower intake of vitamin B3, copper, phosphorus, selenium, sodium, and protein below the recommended dietary allowance may increase the risk of developing IBD. Similarly, reduced intake of vitamin B6, vitamin E, and total PUFA is associated with elevated susceptibility to IBD. CONCLUSION This study elucidates the intricate relationship between nutrient intake and the onset of IBD, underscoring the importance of maintaining a balanced diet for gastrointestinal health. These findings emphasize the significance of informed dietary choices in promoting intestinal wellness and potentially reducing the risk of IBD development.
Collapse
Affiliation(s)
- Nuoya Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Wei Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yajie Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chunlai Nie
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongxia Dan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
93
|
Handa A, Slanzon GS, Ambrosini YM, Haines JM. Effect of Omeprazole on Esophageal Microbiota in Dogs Detected Using a Minimally Invasive Sampling Method. J Vet Intern Med 2025; 39:e70029. [PMID: 40010750 PMCID: PMC11864821 DOI: 10.1111/jvim.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Omeprazole alters the esophageal microbiome (EM) of humans and has associated effects. OBJECTIVES To assess the changes and subsequent recovery of the EM in awake dogs after omeprazole treatment, using the esophageal string test (EST). ANIMALS Ten healthy, client-owned adult dogs. METHODS A prospective longitudinal design was employed, where esophageal samples were initially collected using EST (day 0), involving the oral administration of an EST capsule and subsequent retrieval after 15 min for pH-based segment identification. The dogs were then administered 1 mg/kg of omeprazole orally, twice daily for 14 days. Follow-up EST samplings were conducted on days 15 and 45. Samples were sequenced targeting the V3-V4 region of the 16S rRNA gene and diversity analysis along with differential sequencing (DEseq2) was performed. RESULTS All dogs tolerated the EST without adverse effects. The EST retrieved sufficient biofluid to characterize the EM in this group of dogs. Diversity analysis revealed no significant alterations in alpha (Observed species, Shannon and Simpson indices) and beta diversity (Bray-Curtis) across the time points after omeprazole administration. CONCLUSIONS AND CLINICAL IMPORTANCE Omeprazole therapy did not alter the EM of healthy dogs in this study. The application of EST in dogs illustrates its use as a minimally invasive tool for investigating the role of EM in esophageal health and disease in dogs.
Collapse
Affiliation(s)
- Aditi Handa
- Washington State UniversityPullmanWashingtonUSA
| | | | | | | |
Collapse
|
94
|
Chen X, Yuan Y, Zhou F, Li L, Pu J, Jiang X. m6A RNA methylation: a pivotal regulator of tumor immunity and a promising target for cancer immunotherapy. J Transl Med 2025; 23:245. [PMID: 40022120 PMCID: PMC11871626 DOI: 10.1186/s12967-025-06221-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025] Open
Abstract
M6A modification is one of the most common regulatory mechanisms of gene expression in eukaryotic cells, influencing processes such as RNA splicing, degradation, stability, and protein translation. Studies have shown that m6A methylation is closely associated with tumorigenesis and progression, and it plays a key regulatory role in tumor immune responses. m6A modification participates in regulating the differentiation and maturation of immune cells, as well as related anti-tumor immune responses. In the tumor microenvironment, m6A modification can also affect immune cell recruitment, activation, and polarization, thereby promoting or inhibiting tumor cell proliferation and metastasis, and reshaping the tumor immune microenvironment. In recent years, immunotherapies for tumors, such as immune checkpoint inhibitors and adoptive cell immunotherapy, have been increasingly applied in clinical settings, achieving favorable outcomes. Targeting m6A modifications to modulate the immune system, such as using small-molecule inhibitors to target dysregulated m6A regulatory factors or inducing immune cell reprogramming, can enhance anti-tumor immune responses and strengthen immune cell recognition and cytotoxicity against tumor cells. m6A modification represents a new direction in tumor immunotherapy with promising clinical potential. This review discusses the regulatory role of m6A methylation on immune cells and tumor immune responses and explores new strategies for immunotherapy.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Yixiao Yuan
- Department of Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Fan Zhou
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Lihua Li
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Jun Pu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China.
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China.
| | - Xiulin Jiang
- Department of Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
95
|
Qu X, Ji Y, Long J, Zheng D, Qiao Z, Lin Y, Lu C, Zhou Y, Cheng H. Immuno- and gut microbiota-modulatory activities of β-1,6-glucans from Lentinus edodes. Food Chem 2025; 466:142209. [PMID: 39612846 DOI: 10.1016/j.foodchem.2024.142209] [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: 09/16/2024] [Revised: 11/04/2024] [Accepted: 11/20/2024] [Indexed: 12/01/2024]
Abstract
The β-1,6-glucans from Lentinus edodes have a variety of biological activities. However, the research on extraction and separation of β-1,6-glucans from L. edodes is limited and the yield is low. In the present study, we obtained the high-yield and -purity β-1,6-glucans (ALEPA) from L. edodes by using a sequential extraction and separation process, which is simple and suitable for industrialization. ALEPA significantly promoted the proliferation of splenic T lymphocytes and enhanced the phagocytosis activity of peritoneal macrophages in vivo. 16S rRNA sequencing results showed that ALEPA significantly increased the α diversity of gut microbiota and upregulated the relative abundances of short chain fatty acids (SCFAs)-producing bacterial species. Consistently, the SCFAs in the cecum of mice were upregulated. On a mechanical level, we found that the immunomodulatory effect of ALEPA depended on gut microbiota. Collectively, ALEPA is a promising functional food ingredient that regulates gut microbiota and enhances immune function.
Collapse
Affiliation(s)
- Xian Qu
- Engineering Research Center of Glycoconjugates, Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China
| | - Yahui Ji
- Engineering Research Center of Glycoconjugates, Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China
| | - Jieyi Long
- Infinitus (China) Company Ltd., Guangzhou 510623, China
| | - Donglin Zheng
- Engineering Research Center of Glycoconjugates, Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China
| | - Zhonghui Qiao
- Engineering Research Center of Glycoconjugates, Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China
| | - Yue Lin
- Engineering Research Center of Glycoconjugates, Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China
| | - Chang Lu
- School of Psychology, Northeast Normal University, Changchun 130024, China
| | - Yifa Zhou
- Engineering Research Center of Glycoconjugates, Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China
| | - Hairong Cheng
- Engineering Research Center of Glycoconjugates, Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China..
| |
Collapse
|
96
|
Du D, Li Q, Wei Z, Wang Z, Xu L. Exploring the CDCA-Scd1 Axis: Molecular Mechanisms Linking the Colitis Microbiome to Neurological Deficits. Int J Mol Sci 2025; 26:2111. [PMID: 40076732 PMCID: PMC11900004 DOI: 10.3390/ijms26052111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Inflammatory bowel disease is a risk factor for brain dysfunction; however, the underlying mechanisms remain largely unknown. In this study, we aimed to explore the potential molecular mechanisms through which intestinal inflammation affects brain function and to verify these mechanisms. Mice were treated with multiple cycles of 1% w/v dextran sulfate sodium (DSS) in drinking water to establish a chronic colitis model. Behavioral tests were conducted using the open field test (OFT), tail suspension test (TST), forced swimming test (FST), and Morris water maze test (MWM). Brain metabolomics, transcriptomics, and proteomics analyses were performed, and key target proteins were verified using qPCR and immunofluorescence. Four cycles of DSS administration induced colitis, anxiety, depression, and spatial memory impairment. The integrated multi-omics characterization of colitis revealed decreased brain chenodeoxycholic acid (CDCA) levels as well as reduced stearoyl-CoA desaturase (Scd1) gene and protein expression. Transplantation of the colitis microbiome resulted in anxiety, depression, impaired spatial memory, reduced CDCA content, decreased Scd1 gene and protein expression, and lower concentrations of monounsaturated fatty acids (MUFAs), palmitoleate (C16:1), and oleate (C18:1) in the brain. In addition, CDCA supplementation improved DSS-induced colitis, alleviated depression and spatial memory impairment, and increased Scd1 gene and protein expression as well as MUFA levels in the brain. The gut microbiome induced by colitis contributes to neurological dysfunction, possibly through the CDCA-Scd1 signaling axis. CDCA supplementation alleviates colitis and depressive behavior, likely by increasing Scd1 expression in the brain.
Collapse
Affiliation(s)
- Donglin Du
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; (D.D.)
| | - Qi Li
- Laboratory Animal Center, Chongqing Medical University, Chongqing 400016, China
| | - Zhengqiang Wei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; (D.D.)
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; (D.D.)
| | - Lei Xu
- Laboratory Animal Center, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
97
|
Zhang H, Tian Y, Xu C, Chen M, Xiang Z, Gu L, Xue H, Xu Q. Crosstalk between gut microbiotas and fatty acid metabolism in colorectal cancer. Cell Death Discov 2025; 11:78. [PMID: 40011436 DOI: 10.1038/s41420-025-02364-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/30/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common malignancy globally and the second leading cause of cancer-related mortality. Its development is a multifactorial and multistage process influenced by a dynamic interplay between gut microbiota, environmental factors, and fatty acid metabolism. Dysbiosis of intestinal microbiota and abnormalities in microbiota-associated metabolites have been implicated in colorectal carcinogenesis, highlighting the pivotal role of microbial and metabolic interactions. Fatty acid metabolism serves as a critical nexus linking dietary patterns with gut microbial activity, significantly impacting intestinal health. In CRC patients, reduced levels of short-chain fatty acids (SCFAs) and SCFA-producing bacteria have been consistently observed. Supplementation with SCFA-producing probiotics has demonstrated tumor-suppressive effects, while therapeutic strategies aimed at modulating SCFA levels have shown potential in enhancing the efficacy of radiation therapy and immunotherapy in both preclinical and clinical settings. This review explores the intricate relationship between gut microbiota, fatty acid metabolism, and CRC, offering insights into the underlying mechanisms and their potential translational applications. Understanding this interplay could pave the way for novel diagnostic, therapeutic, and preventive strategies in the management of CRC.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Yuan Tian
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Chunjie Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Miaomiao Chen
- Department of Radiology, Huashan Hospital, National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200040, PR China
| | - Zeyu Xiang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Lei Gu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Hanbing Xue
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Qing Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
98
|
Sigall Boneh R, van der Kruk N, Wine E, Verburgt CM, de Meij TGJ, Löwenberg M, Gecse KB, Wierdsma N, Derikx JPM, de Jonge WJ, D’Haens G, Ghiboub M, Van Limbergen JE. Tryptophan metabolites profile predict remission with dietary therapy in pediatric Crohn's disease. Therap Adv Gastroenterol 2025; 18:17562848251323004. [PMID: 40012837 PMCID: PMC11863242 DOI: 10.1177/17562848251323004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
Background Crohn's disease (CD) exclusion diet combined with partial enteral nutrition (CDED + PEN) or exclusive enteral nutrition (EEN) is effective in inducing remission in mild-to-moderate pediatric CD. Although CDED + PEN is better tolerated and has higher compliance compared to EEN, a subset of patients does not achieve remission. Diet-induced remission is shown to be positively associated with specific changes in tryptophan-metabolites. Objectives To investigate whether the abundance of baseline fecal tryptophan-metabolites predicts dietary therapy outcomes in pediatric CD. Design Diagnostic accuracy study and secondary analysis of previously conducted Randomized Controlled Trial (RCT). Methods Twenty-six patients from previously performed RCT of mild-to-moderate pediatric CD were included. The patients were classified as having clinical remission (R) (n = 19 in total; CDED + PEN = 10 and to EEN = 9) or No-Remission (NR) (n = 7 in total; CDED + PEN = 3 and EEN = 4) following 6 weeks of therapy, based on the Pediatric Crohn's Disease Activity Index score (⩽10 = remission). We performed a targeted quantitative analysis of 21 tryptophan-metabolites in baseline (t = 0) fecal samples from both groups, utilizing liquid chromatography coupled with quadrupole mass spectrometry. Receiver operator characteristic curve (ROC) and random forest analysis (RFA) were used to assess the predictive power of fecal tryptophan-metabolites for dietary outcomes at baseline. Ratios of tryptophan-metabolites were compared to investigate different downstream tryptophan pathways. Results Baseline fecal kynurenine level was significantly higher in NR compared to R for CDED + PEN (p = 0.02) and EEN (p = 0.04). ROC analysis highlighted the robust predictive power of kynurenine for CDED + PEN (area under the curve (AUC = 0.97)) and EEN (AUC = 0.88)-induced remission. RFA corroborated these observations. The ratio serotonin/kynurenine was the strongest predictor of CDED + PEN-induced remission (AUC = 1). The ratio 5-hydroxytryptophan/kynurenine (AUC = 0.88) predicted EEN-induced remission. By combining data from CDED + PEN and EEN, kynurenine (AUC = 0.91) and ratios of quinolinic acid/kynurenine (AUC = 0.93) and kynurenine/indole-3-acetic acid (AUC = 0.88) demonstrated strong predictive performance for dietary therapy-induced remission. Conclusion Baseline tryptophan metabolites have the potential to serve as a biomarker for dietary remission in pediatric CD. Some tryptophan metabolite ratios showed the most promising predictive capabilities. If confirmed in validation studies, baseline fecal tryptophan markers may be able to provide much-needed guidance to personalize dietary intervention within the management of pediatric CD. Trial registration NCT01728870.
Collapse
Affiliation(s)
- Rotem Sigall Boneh
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Pediatric Gastroenterology and Nutrition Unit, The E. Wolfson Medical Center, Holon, Israel
| | - Nikki van der Kruk
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Eytan Wine
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Charlotte M. Verburgt
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Tim G. J. de Meij
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Mark Löwenberg
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Krisztina B. Gecse
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Nicolette Wierdsma
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Joep P. M. Derikx
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Wouter J. de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Department of Surgery, University of Bonn, Bonn, Germany
| | - Geert D’Haens
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Mohammed Ghiboub
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Meibergdreef 69, Amsterdam 1105 BK, The Netherlands
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Johan E. Van Limbergen
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
99
|
De Jaegher S, D’Aguanno M, Pinzauti D, Biazzo M. Nasal Microbiota Profiling as a Predictive Secondary Tool for COVID-19 Diagnosis: The Critical Role of Taxonomic Resolution. Microorganisms 2025; 13:501. [PMID: 40142394 PMCID: PMC11944269 DOI: 10.3390/microorganisms13030501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
The SARS-CoV-2 pandemic has led to an urgent need for effective and rapid diagnostic tools. In the present study, we have evaluated the predictive diagnostic potential of nasal microbiota by analyzing microbial community structures at different taxonomic level resolutions-species, genus, family, order, class and phylum-using Random Forest modelling. A total of 179 nasal swabs from COVID-19-positive (n = 85) and COVID-19-negative (n = 94) individuals were sequenced using a full-length 16S rRNA sequencing (Oxford Nanopore) approach. During each iteration of the Random Forest model, the dataset was randomly split into a training set (70%) and a testing set (30%). Model performance improved with finer taxonomic resolution, achieving the highest accuracy at the Species level (AUROC = 0.821 ± 0.059) and a sensitivity of 55.6% at a specificity threshold of 90%. A progressive decline in AUROC and sensitivity was observed at broader taxonomic levels. Furthermore, Beta diversity analysis supported that microbial community structures are more distinct between COVID-19-positive and COVID-19-negative groups at finer taxonomic levels. These findings highlight the potential role of nasal microbiota profiling as a secondary diagnostic tool for COVID-19, particularly at species- and genus-level classification, and underscore the importance of high taxonomic resolution in microbiome-based diagnostics. However, limited by an uneven sample distribution and the lack of medical evaluations, further large-scale studies are needed before the nasal microbiota can be implemented in the clinical diagnostics of COVID-19.
Collapse
Affiliation(s)
- Simon De Jaegher
- The BioArte Ltd., Life Science Park, Triq San Gijan, 3000 San Gwann, Malta; (S.D.J.); (M.D.); (M.B.)
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Maria D’Aguanno
- The BioArte Ltd., Life Science Park, Triq San Gijan, 3000 San Gwann, Malta; (S.D.J.); (M.D.); (M.B.)
| | - David Pinzauti
- The BioArte Ltd., Life Science Park, Triq San Gijan, 3000 San Gwann, Malta; (S.D.J.); (M.D.); (M.B.)
| | - Manuele Biazzo
- The BioArte Ltd., Life Science Park, Triq San Gijan, 3000 San Gwann, Malta; (S.D.J.); (M.D.); (M.B.)
| |
Collapse
|
100
|
Shi T, Zhang H, Chen Y. The m6A revolution: transforming tumor immunity and enhancing immunotherapy outcomes. Cell Biosci 2025; 15:27. [PMID: 39987091 PMCID: PMC11846233 DOI: 10.1186/s13578-025-01368-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/14/2025] [Indexed: 02/24/2025] Open
Abstract
N6-methyladenosine (m6A), the most prevalent RNA modification in eukaryotes, plays a critical role in the development and progression of various diseases, including cancer, through its regulation of RNA degradation, stabilization, splicing, and cap-independent translation. Emerging evidence underscores the significant role of m6A modifications in both pro-tumorigenic and anti-tumorigenic immune responses. In this review, we provide a comprehensive overview of m6A modifications and examine the relationship between m6A regulators and cancer immune responses. Additionally, we summarize recent advances in understanding how m6A modifications influence tumor immune responses by directly modulating immune cells (e.g., dendritic cells, tumor-associated macrophages, and T cells) and indirectly affecting cancer cells via mechanisms such as cytokine and chemokine regulation, modulation of cell surface molecules, and metabolic reprogramming. Furthermore, we explore the potential synergistic effects of targeting m6A regulators in combination with immune checkpoint inhibitor (ICI) therapies. Together, this review consolidates current knowledge on the role of m6A-mediated regulation in tumor immunity, offering insights into how a deeper understanding of these modifications may identify patients who are most likely to benefit from immunotherapies.
Collapse
Affiliation(s)
- Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
| | - Huan Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
| | - Yueqiu Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
| |
Collapse
|