1
|
Geng P, Zhao N, Zhou Y, Harris RS, Ge Y. Faecalibacterium prausnitzii regulates carbohydrate metabolic functions of the gut microbiome in C57BL/6 mice. Gut Microbes 2025; 17:2455503. [PMID: 39841201 DOI: 10.1080/19490976.2025.2455503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/08/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
The probiotic impact of microbes on host metabolism and health depends on both host genetics and bacterial genomic variation. Faecalibacterium prausnitzii is the predominant human gut commensal emerging as a next-generation probiotic. Although this bacterium exhibits substantial intraspecies diversity, it is unclear whether genetically distinct F. prausnitzii strains might lead to functional differences in the gut microbiome. Here, we isolated and characterized a novel F. prausnitzii strain (UT1) that belongs to the most prevalent but underappreciated phylogenetic clade in the global human population. Genome analysis showed that this butyrate-producing isolate carries multiple putative mobile genetic elements, a clade-specific defense system, and a range of carbohydrate catabolic enzymes. Multiomic approaches were used to profile the impact of UT1 on the gut microbiome and associated metabolic activity of C57BL/6 mice at homeostasis. Both 16S rRNA and metagenomic sequencing demonstrated that oral administration of UT1 resulted in profound microbial compositional changes including a significant enrichment of Lactobacillus, Bifidobacterium, and Turicibacter. Functional profiling of the fecal metagenomes revealed a markedly higher abundance of carbohydrate-active enzymes (CAZymes) in UT1-gavaged mice. Accordingly, UT1-conditioned microbiota possessed the elevated capability of utilizing starch in vitro and exhibited a lower availability of microbiota-accessible carbohydrates in the gut. Further analysis uncovered a functional network wherein UT1 reduced the abundance of mucin-degrading CAZymes and microbes, which correlated with a concomitant reduction of fecal mucin glycans. Collectively, our results reveal a crucial role of UT1 in facilitating the carbohydrate metabolism of the gut microbiome and expand our understanding of the genetic and phenotypic diversity of F. prausnitzii.
Collapse
Affiliation(s)
- Peiling Geng
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Ni Zhao
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yufan Zhou
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Reuben S Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yong Ge
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
2
|
Rahaman MM, Wangchuk P, Sarker S. A systematic review on the role of gut microbiome in inflammatory bowel disease: Spotlight on virome and plant metabolites. Microb Pathog 2025; 205:107608. [PMID: 40250496 DOI: 10.1016/j.micpath.2025.107608] [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: 08/02/2024] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Inflammatory bowel diseases (IBD), including ulcerative colitis and Crohn's disease, arise from various factors such as dietary, genetic, immunological, and microbiological influences. The gut microbiota plays a crucial role in the development and treatment of IBD, though the exact mechanisms remain uncertain. Current research has yet to definitively establish the beneficial effects of the microbiome on IBD. Bacteria and viruses (both prokaryotic and eukaryotic) are key components of the microbiome uniquely related to IBD. Numerous studies suggest that dysbiosis of the microbiota, including bacteria, viruses, and bacteriophages, contributes to IBD pathogenesis. Conversely, some research indicates that bacteria and bacteriophages may positively impact IBD outcomes. Additionally, plant metabolites play a crucial role in alleviating IBD due to their anti-inflammatory and microbiome-modulating properties. This systematic review discusses the role of the microbiome in IBD pathogenesis and evaluates the potential connection between plant metabolites and the microbiome in the context of IBD pathophysiology.
Collapse
Affiliation(s)
- Md Mizanur Rahaman
- Biomedical Sciences and Molecular Biology, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, 4811, Australia
| | - Phurpa Wangchuk
- College of Science and Engineering, James Cook University, Nguma Bada campus, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Nguma Bada campus, McGregor Rd, Smithfield, Cairns, QLD, 4878, Australia
| | - Subir Sarker
- Biomedical Sciences and Molecular Biology, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, 4811, Australia.
| |
Collapse
|
3
|
Xu J, Xu H, Li J, Huang W, Li Y, Guo X, Zhu M, Peng Y, Zhou Y, Nie Y. Clostridium butyricum-induced balance in colonic retinol metabolism and short-chain fatty acid levels inhibit IgA-related mucosal immunity and relieve colitis developments. Microbiol Res 2025; 298:128203. [PMID: 40319662 DOI: 10.1016/j.micres.2025.128203] [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: 12/06/2024] [Revised: 04/08/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Gut microbiota and their metabolites play a significant role in inflammatory bowel disease. Here, we attempted to determine the anti-inflammatory role of the probiotic Clostridium. butyricum (CB) in inflammatory bowel disease and identify the exact immune mechanism. The clinical significance of Clostridiales and CB was explored in patients with ulcerative colitis. The inflammation-suppressive role of CB was evaluated in mice with DSS-induced colitis. 16S rRNA sequencing was performed to assess changes in the gut microbiota. Altered transcription levels were detected by RNA sequencing. Flow cytometry was performed to assess the frequency of IgA responses to gut microbiota. Clostridiales and CB were depleted in ulcerative colitis. Oral gavage with CB significantly suppressed weight loss and colon shortening in the dextran sulfate sodium-induced colitis mouse model. Intestinal barrier injury was reversed and the gut microbiota was restored upon treatment with CB administration. The mucosal immune response to gut microbiota was reversed upon treatment with CB. CB conditional medium was more effective than heat-killed CB in alleviating inflammation. Mechanistically, retinol metabolism and retinoic acid levels were higher in groups treated with CB and butyrate. CB and the metabolite butyrate exerted a suppressive role on the abundance of Immunoglobulin A-coated gut microbiota by inhibiting retinoic acid synthesis. In summary, CB protects against inflammation and intestinal barrier injury by producing anti-inflammatory metabolites that can regulate the mucosal immune response to gut microbiota by increasing retinoic acid levels in the colon.
Collapse
Affiliation(s)
- Jing Xu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, China; Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Haoming Xu
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Jianhong Li
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, China; Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Wenqi Huang
- Division of Rheumatology, Department of Medicine/Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Yingfei Li
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Xue Guo
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, China; Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Minzheng Zhu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, China; Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Yao Peng
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.
| | - Youlian Zhou
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.
| | - Yuqiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Ma H, Li R, Qu B, Liu Y, Li P, Zhao J. The Role of Bile Acid in Immune-Mediated Skin Diseases. Exp Dermatol 2025; 34:e70108. [PMID: 40302108 DOI: 10.1111/exd.70108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 04/07/2025] [Accepted: 04/19/2025] [Indexed: 05/01/2025]
Abstract
Immune-mediated skin disorders arise from dysfunctional immune responses, instigating inflammatory dermatoses and a reduced quality of life. The complex pathogenesis likely involves genetic risks, environmental triggers and aberrant immune activation. An emerging body of evidence suggests that bile acid disturbances may critically promote immune pathology in certain skin conditions. Bile acids synthesised from cholesterol regulate nutrient metabolism and immune cell function via nuclear receptors and G protein-coupled receptors (GPCRs). Altered bile acid profiles and receptor expression have been identified in psoriasis, atopic dermatitis (AD) and autoimmune blistering diseases. Disruptions in bile acid signalling affect the inflammatory and metabolic pathways linked to these disorders. Targeting components of the bile acid axis represents a promising therapeutic strategy. This review elucidates the intricate links between bile acid homeostasis and immune dysfunction in inflammatory skin diseases, synthesising evidence that targeting bile acid pathways may unlock innovative therapeutic avenues. This study compiles clinical and experimental data revealing disrupted bile acid signalling and composition in various immune-mediated dermatoses, highlighting the emerging significance of bile acids in cutaneous immune regulation.
Collapse
Affiliation(s)
- Huike Ma
- Beijing Hospital of Traditional Chinese Medicine, Beijing Institute of Chinese Medicine, Capital Medical University, Beijing, China
| | - Ruonan Li
- Beijing Hospital of Traditional Chinese Medicine, Beijing Institute of Chinese Medicine, Capital Medical University, Beijing, China
| | - Baoquan Qu
- Beijing Hospital of Traditional Chinese Medicine, Beijing Institute of Chinese Medicine, Capital Medical University, Beijing, China
| | - Yuchen Liu
- Beijing Hospital of Traditional Chinese Medicine, Beijing Institute of Chinese Medicine, Capital Medical University, Beijing, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Beijing Institute of Chinese Medicine, Capital Medical University, Beijing, China
| | - Jingxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Beijing Institute of Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Hirasaki M, Kadowaki R, Xuan AAL, Harata G, Miyazawa K, Maeno S, Gueimonde M, Endo A. Species-level quantification of Faecalibacterium spp. in faeces of healthy Japanese adults. J Med Microbiol 2025; 74:002019. [PMID: 40408134 PMCID: PMC12102495 DOI: 10.1099/jmm.0.002019] [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: 03/16/2025] [Accepted: 05/08/2025] [Indexed: 05/25/2025] Open
Abstract
Faecalibacterium prausnitzii has been considered one of the predominant microbes in the gut microbiota of healthy human adults. Moreover, due to its beneficial metabolites and its reduced population in patients with various disorders, this organism has been regarded as one of the key gut microbes in human health. However, following recent revisions in the taxonomy of the genus Faecalibacterium and F. prausnitzii, the reported population distribution and health benefits of this species have become unclear. In the present study, the population of nine species-level taxonomic groups (hereafter referred to as species) within Faecalibacterium was quantified at the species level in the faeces of healthy Japanese adults (n=88). qPCR, combined with rpoA-based species-specific primers, showed that Faecalibacterium taiwanense had the highest detection rate (prevalence) and copy number among Faecalibacterium spp., followed by Faecalibacterium longum, Faecalibacterium duncaniae and F. prausnitzii, while the remaining five species were detected only occasionally. The population of F. duncaniae varied significantly between age groups, being higher in individuals in their 40s and 50s compared to those in their 20s (P=0.047 and 0.002, respectively). The present study indicates that F. prausnitzii is not the predominant Faecalibacterium species in the healthy Japanese adults included in the present study. Future studies will shed light on the health benefits of the dominant Faecalibacterium spp.
Collapse
Affiliation(s)
- Masahiro Hirasaki
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, 156-8502 Tokyo, Japan
| | - Ren Kadowaki
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., 241-0021 Kanagawa, Japan
| | - Adeline Ang Li Xuan
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, 156-8502 Tokyo, Japan
| | - Gaku Harata
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., 241-0021 Kanagawa, Japan
| | - Kenji Miyazawa
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., 241-0021 Kanagawa, Japan
| | - Shintaro Maeno
- Research Center for Advanced Science and Innovation, Organization for Research Initiatives, Yamaguchi University, 753-8515 Yamaguchi, Japan
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, IPLA-CSIC, Paseo Rio Linares s/n, 33300 Villaviciosa, Spain
| | - Akihito Endo
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, 156-8502 Tokyo, Japan
| |
Collapse
|
6
|
Petracco G, Faimann I, Reichmann F. Inflammatory bowel disease and neuropsychiatric disorders: Mechanisms and emerging therapeutics targeting the microbiota-gut-brain axis. Pharmacol Ther 2025; 269:108831. [PMID: 40023320 DOI: 10.1016/j.pharmthera.2025.108831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/03/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are the two major entities of inflammatory bowel disease (IBD). These disorders are known for their relapsing disease course and severe gastrointestinal symptoms including pain, diarrhoea and bloody stool. Accumulating evidence suggests that IBD is not only restricted to the gastrointestinal tract and that disease processes are able to reach distant organs including the brain. In fact, up to 35 % of IBD patients also suffer from neuropsychiatric disorders such as generalized anxiety disorder and major depressive disorder. Emerging research in this area indicates that in many cases these neuropsychiatric disorders are a secondary condition as a consequence of the disturbed communication between the gut and the brain via the microbiota-gut-brain axis. In this review, we summarise the current knowledge on IBD-associated neuropsychiatric disorders. We examine the role of different pathways of the microbiota-gut-brain axis in the development of CNS disorders highlighting altered neural, immunological, humoral and microbial communication. Finally, we discuss emerging therapies targeting the microbiota-gut-brain axis to alleviate IBD and neuropsychiatric symptoms including faecal microbiota transplantation, psychobiotics, microbial metabolites and vagus nerve stimulation.
Collapse
Affiliation(s)
- Giulia Petracco
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Isabella Faimann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Florian Reichmann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria; BiotechMed-Graz, Austria.
| |
Collapse
|
7
|
Li S, Niu XX, Liu JL, Su M, Li QQ, Wang CY, Wang JJ, Chen HY, Ji D. Leveraging the gut microbiome to understand the risk factor of cognitive impairment in patients with liver cirrhosis. Eur J Gastroenterol Hepatol 2025; 37:627-637. [PMID: 39976005 DOI: 10.1097/meg.0000000000002934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
OBJECTIVES The role of the gut-liver axis in liver cirrhosis is becoming increasingly recognized. We investigated the fecal microbiome in patients with liver cirrhosis and its potential function as a predictive biomarker of hepatic encephalopathy. METHODS Patients were divided into either a high plasma ammonia (HPA) group or a low plasma ammonia (LPA) group according to the upper limit of normal of plasma ammonia concentration. 16S rRNA sequencing of fecal samples was performed to study how the microbiota affects the clinical symptoms of liver cirrhosis. The Stroop test was used to assess the ability of the brain to inhibit habitual behaviors. RESULTS Totally, 21 subjects were enrolled. Among the 18 patients with liver cirrhosis, 14 were male, the age range was 42-56 years, and the plasma ammonia level range was 20-125.9 μmol/l. The Stroop test showed more severe cognitive impairment in HPA than in LPA individuals. At the same time, there were significant differences in fecal microbiome characteristics between the two groups, characterized by a further increase in the abundance of the Proteobacteria phylum in the gut (especially aerobic Enterobacteriaceae ). Function predictions of Phylogenetic Investigation of Communities by Reconstruction of Unobserved States in the microbiome further explained the increase in the Enterobacteriaceae -dominated polyamine synthesis pathway in the gut microbiome of HPA groups. CONCLUSION Cirrhotic patients with hyperammonemia have a specific fecal bacterial composition (characterized via expansion of Enterobacteriaceae ). The ability to bio-synthesize polyamines that Enterobacteriaceae possesses is likely to be a key factor in the elevation of plasma ammonia.
Collapse
Affiliation(s)
- Shuyao Li
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| | - Xiao-Xia Niu
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| | - Jia-Liang Liu
- Department of General Internal Medicine, Hospital of North China Electric Power University, Beijing, China
| | - Min Su
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| | - Qian-Qian Li
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| | - Chun-Yan Wang
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| | - Jian-Jun Wang
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| | - Hong-Yan Chen
- Department of General Internal Medicine, Hospital of North China Electric Power University, Beijing, China
| | - Dong Ji
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| |
Collapse
|
8
|
Mclellan P, Auger S, Goudiaby MT, Brot L, Benech N, Grill JP, Bourrier A, Mariat D, Mayeur C, Thomas M, Robert V, Kirchgesner J, Beaugerie L, Sokol H, Langella P, Seksik P, Chatel JM. Faecalibacterium Diversity in the Gut Microbiome of Crohn's Disease Patients. United European Gastroenterol J 2025. [PMID: 40252217 DOI: 10.1002/ueg2.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 04/21/2025] Open
Abstract
Faecalibacterium has recently garnered attention for its potential health implications. To better understand its role, we developed and assessed real-time PCR assays for detecting and quantifying various Faecalibacterium species in human stool samples from both healthy individuals and Crohn's disease patients, either in flare or remission. The assays targeted the Microbial Anti-inflammatory Molecule (MAM) genes, which encode MAM proteins. These assays demonstrated 100% species-specificity using strains from six Faecalibacterium species: Faecalibacterium prausnitzii, Faecalibacterium taiwanense, Faecalibacterium duncaniae, Faecalibacterium longum, Faecalibacterium hattori, and Faecalibacterium CNCM4541. They also showed high sensitivity with detection limits of 10^5 bacteria per gram of sample. In healthy individuals, the different Faecalibacterium species varied in abundance. F. taiwanense, F. duncaniae, and F. longum were the most prevalent, around 10^10 bacteria/g of stool. In contrast, F. hattori and CNCM4541 were less abundant, with 10^7 bacteria/g. Despite its low abundance, F. hattori was present in all healthy subjects, while CNCM4541 was detected in only 50% of them. Notably, F. taiwanense, F. duncaniae, and F. longum were found in all healthy individuals. In Crohn's disease patients, both in flare and remission, a decrease in Faecalibacterium species was observed, with no recovery in remission. The most abundant species in Crohn's disease patients were F. prausnitzii and F. duncaniae, around 10^7 bacteria/g, while F. longum, F. hattori, and F. taiwanense were present at lower levels (10^6 bacteria/g), and CNCM4541 was no longer detected. Interestingly, F. prausnitzii showed a smaller decrease in abundance compared with other species. Moreover, F. prausnitzii was significantly more prevalent in patients in remission than in those in flare, suggesting that it may be more resistant to inflammation. These findings highlight the importance of accurately characterizing and quantifying Faecalibacterium species to better understand their role in health and disease.
Collapse
Affiliation(s)
- Paul Mclellan
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, Inserm, Paris, France
| | - Sandrine Auger
- Université Paris-Saclay, INRAE, AgroParisTech, UMR 1319 Micalis, Jouy-en-Josas, France
| | | | - Loic Brot
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, Inserm, Paris, France
| | - Nicolas Benech
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, Inserm, Paris, France
| | - Jean Pierre Grill
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, Inserm, Paris, France
| | - Anne Bourrier
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, Inserm, Paris, France
| | - Denis Mariat
- Université Paris-Saclay, INRAE, AgroParisTech, UMR 1319 Micalis, Jouy-en-Josas, France
| | - Camille Mayeur
- Université Paris-Saclay, INRAE, AgroParisTech, UMR 1319 Micalis, Jouy-en-Josas, France
| | - Muriel Thomas
- Université Paris-Saclay, INRAE, AgroParisTech, UMR 1319 Micalis, Jouy-en-Josas, France
| | - Véronique Robert
- Université Paris-Saclay, INRAE, AgroParisTech, UMR 1319 Micalis, Jouy-en-Josas, France
| | - Julien Kirchgesner
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, Inserm, Paris, France
| | - Laurent Beaugerie
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, Inserm, Paris, France
| | - Harry Sokol
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, Inserm, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Philippe Langella
- Université Paris-Saclay, INRAE, AgroParisTech, UMR 1319 Micalis, Jouy-en-Josas, France
| | - Philippe Seksik
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, Inserm, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Jean Marc Chatel
- Université Paris-Saclay, INRAE, AgroParisTech, UMR 1319 Micalis, Jouy-en-Josas, France
| |
Collapse
|
9
|
Bai R, Ma L, Li F, Pan L, Bao Y, Li X, Wang S, Yue H, Zheng F. Total ginsenosides from wild ginseng improve immune regulation in a rat model of spleen qi deficiency by modulating fecal-bacteria-associated short-chain fatty acids and intestinal barrier integrity. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1256:124554. [PMID: 40081219 DOI: 10.1016/j.jchromb.2025.124554] [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: 01/13/2025] [Revised: 01/17/2025] [Accepted: 03/01/2025] [Indexed: 03/15/2025]
Abstract
For thousands of years, traditional Chinese medicine (TCM) has made extensive use of wild ginseng. It is thought to provide vital energy effects and to boost immunity. This study aimed to clarify the processes by which short-chain fatty acids (SCFAs) metabolites and the intestinal barrier are used by total ginsenosides wild ginseng (TWG) to modulate immunity. In this study, we analyzed and identified ginsenosides in the colon using UPLC-Q-TOF-MSE methods. In the meantime, a rat model of spleen qi deficiency (SQD) was created using reserpine, and the effects of TWG on intestinal barrier function and short-chain fatty acids in the feces of SQD-affected rats were examined. 28 ginsenosides were found in the colon during this experiment, and the main components were measured. TWG considerably increased fecal concentrations of acetic, propionic and 6 others, according to SCFAs analysis. According to serum immunological markers, TWG reduced IL-17 and IL-1β levels, increased IL-10, IL-22, and TGF-β concentrations, balanced Th17/Treg ratios, and reduced toxicants such DAO and LPS in rats with SQD. TWG improved barrier function, reduced permeability, increased tight junction protein expression, and lessened intestinal injury. A favorable correlation between intestinal barrier proteins and fatty acids was shown by correlation studies. The gut barrier and SCFAs perspectives helped to clarify the mechanism by which TWG controls immune activity. This study offers a fresh theoretical framework for TWG's future advancement and application.
Collapse
Affiliation(s)
- Ruobing Bai
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Liting Ma
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Fangtong Li
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Lijia Pan
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Yuwen Bao
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Xinze Li
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Shen Wang
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Hao Yue
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China.
| | - Fei Zheng
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China.
| |
Collapse
|
10
|
El Boukhari R, Matin M, Bouissane L, Ławiński M, Lushchak O, Singla RK, Mickael M, Mayneris‐Perxachs J, Grafakou ME, Xu S, Liu B, Guan J, Półtorak A, Szpicer A, Wierzbicka A, Tzvetkov NT, Banach M, Horbańczuk JO, Jóźwik A, Cascella M, Shen B, Pirgozliev VR, Wang D, Litvinova O, Adamska O, Kamińska A, Łapiński M, Stolarczyk A, Berindan‐Neagoe I, Milella L, Yeung AWK, Suravajhala P, Bishayee A, Lordan R, Iantovics LB, Lagoa R, Michalczuk M, Stoyanov J, Kinghorn AD, Jalil B, Weckwerth W, Goh BH, Li M, Chaubey G, Russo GL, Frazzini S, Rossi L, Battino M, Jia W, Su Q, Ma X, Rollinger JM, Rittmann SKR, Sheridan H, Walsh JJ, Lizard G, Karpiński TM, Silva AS, Piwowarski J, Xie L, Fan T, Giampieri F, El Midaoui A, Wong K, Gan R, Fatimi A, Atanasov AG. Enhancing human gut health: Global innovations in dysbiosis management. IMETA 2025. [DOI: 10.1002/imt2.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/26/2025] [Indexed: 05/03/2025]
Affiliation(s)
- Reda El Boukhari
- Chemical Science and Engineering Research Team (ERSIC), Department of Chemistry, Polydisciplinary Faculty of Beni Mellal (FPBM) Sultan Moulay Slimane University (USMS) Beni Mellal Morocco
| | - Maima Matin
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Latifa Bouissane
- Molecular Chemistry, Materials and Catalysis Laboratory, Faculty of Sciences and Technologies Sultan Moulay Slimane University Beni Mellal Morocco
| | - Michał Ławiński
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
- Department of General, Gastroenterologic and Oncologic Surgery Medical University of Warsaw Warsaw Poland
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology Vasyl Stefanyk Precarpathian National University Ivano‐Frankivsk Ukraine
- Research and Development University Ivano‐Frankivsk Ukraine
| | - Rajeev K. Singla
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu Sichuan China
- School of Pharmaceutical Sciences Lovely Professional University Phagwara Punjab India
| | - Michel‐Edwar Mickael
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Jordi Mayneris‐Perxachs
- Department of Diabetes, Endocrinology and Nutrition Dr. Josep Trueta University Hospital Girona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) Madrid Spain
- Integrative Systems Medicine and Biology Group, Girona Biomedical Research Institute (IDIBGI‐CERCA) Parc Hospitalari Martí i Julià Salt Spain
| | - Maria Eleni Grafakou
- Chair of Pharmaceutical Biology, Faculty of Pharmacy and Chemistry University of Regensburg Germany
| | - Shuhua Xu
- Center for Evolutionary Biology, School of Life Sciences Fudan University Shanghai China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center Fudan University Shanghai China
| | - Bowen Liu
- School of Agriculture Yunnan University Kunming China
| | - Jiayi Guan
- Henan Institute of Medical and Pharmaceutical Sciences Zhengzhou University Zhengzhou China
| | - Andrzej Półtorak
- Department of Technique and Food Development, Institute of Human Nutrition Sciences Warsaw University of Life Sciences Warsaw Poland
| | - Arkadiusz Szpicer
- Department of Technique and Food Development, Institute of Human Nutrition Sciences Warsaw University of Life Sciences Warsaw Poland
| | - Agnieszka Wierzbicka
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
- Department of Technique and Food Development, Institute of Human Nutrition Sciences Warsaw University of Life Sciences Warsaw Poland
| | - Nikolay T. Tzvetkov
- Department of Biochemical Pharmacology and Drug Design, Institute of Molecular Biology “Roumen Tsanev” Bulgarian Academy of Sciences Sofia Bulgaria
| | - Maciej Banach
- Faculty of Medicine The John Paul II Catholic University of Lublin (KUL) Lublin Poland
- Department of Cardiology and Adult Congenital Heart Diseases Polish Mother's Memorial Hospital Research Institute (PMMHRI) Lodz Poland
- Department of Preventive Cardiology and Lipidology Medical University of Lodz (MUL) Lodz Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Jarosław Olav Horbańczuk
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Artur Jóźwik
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Marco Cascella
- Anesthesia and Pain Medicine, Department of Medicine, Surgery and Dentistry “Scuola MedicaSalernitana” University of Salerno Baronissi Italy
| | - Bairong Shen
- Department of Critical Care Medicine and Institutes for Systems Genetics Frontiers Science Center for Disease‐Related Molecular Network, West China Hospital,Sichuan University Chengdu Sichuan China
- Center for High Altitude Medicine, West China Hospital Sichuan University Chengdu Sichuan China
| | | | - Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research McMaster University Hamilton Ontario Canada
- Division of Endocrinology and Metabolism, Department of Medicine McMaster University Hamilton Ontario Canada
| | - Olena Litvinova
- National University of Pharmacy of the Ministry of Health of Ukraine Kharkiv Ukraine
- Ludwig Boltzmann Institute Digital Health and Patient Safety Medical University of Vienna Vienna Austria
| | - Olga Adamska
- Faculty of Medicine Collegium Medicum Cardinal Stefan Wyszyński University in Warsaw Warsaw Poland
| | - Agnieszka Kamińska
- Faculty of Medicine Collegium Medicum Cardinal Stefan Wyszyński University in Warsaw Warsaw Poland
| | - Marcin Łapiński
- Orthopaedic and Rehabilitation Department Medical University of Warsaw Warsaw Poland
| | - Artur Stolarczyk
- Orthopaedic and Rehabilitation Department Medical University of Warsaw Warsaw Poland
| | - Ioana Berindan‐Neagoe
- Department of Genomics MEDFUTURE ‐ Institute for Biomedical Research“Iuliu Hațieganu” University of Medicine and Pharmacy No. 23 Cluj‐Napoca Romania
| | - Luigi Milella
- Department of Health Sciences University of Basilicata Potenza Italy
| | - Andy Wai Kan Yeung
- Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry The University of Hong Kong Pokfulam Hong Kong SAR
| | - Prashanth Suravajhala
- Amrita School of Biotechnology Amrita Viswa Vidyapeetham Clappana Kerala India
- Department of Biosciences Manipal University Jaipur, Dehmi Kala Jaipur Rajasthan India
| | - Anupam Bishayee
- Department of Pharmacology College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine Bradenton Florida USA
| | - Ronan Lordan
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| | - Laszlo Barna Iantovics
- Department of Electrical Engineering and Information Technology George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures Targu Mures Romania
| | - Ricardo Lagoa
- ESTG‐Polytechnic Institute of Leiria Morro do Lena‐Alto do Vieiro Leiria Portugal
- LSRE‐LCM‐Associate Laboratory in Chemical Engineering University of Porto Porto Portugal
| | - Monika Michalczuk
- Department of Animal Breeding, Institute of Animal Sciences Warsaw University of Life Sciences ‐ SGGW Warsaw Poland
| | - Jivko Stoyanov
- Swiss Paraplegic Research Nottwil Switzerland
- Institute of Social and Preventive Medicine (ISPM) University of Bern Bern Switzerland
| | | | - Banaz Jalil
- Pharmacognosy and Phytotherapy UCL School of Pharmacy London UK
| | - Wolfram Weckwerth
- Molecular Systems Biology Lab (MOSYS), Department of Functional and Evolutionary Ecology University of Vienna Vienna Austria
- Vienna Metabolomics Center (VIME) University of Vienna Vienna Austria
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre (SBMDC) School of Medical and Life Sciences Subang Jaya Malaysia
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy Monash University Malaysia Subang Jaya Malaysia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine University of Technology Sydney Ultimo New South Wales Australia
| | - Meng‐Yao Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
- Department of Biliary‐Pancreatic Surgery, Renji Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Gyaneshwer Chaubey
- Cytogenetics Laboratory, Department of Zoology Banaras Hindu University Varanasi Uttar Pradesh India
| | - Gian Luigi Russo
- National Research Council Institute of Food Sciences Avellino Italy
| | - Sara Frazzini
- Department of Veterinary Medicine and Animal Science (DIVAS) University of Milan Lodi Italy
| | - Luciana Rossi
- Department of Veterinary Medicine and Animal Science (DIVAS) University of Milan Lodi Italy
| | - Maurizio Battino
- Department of Clinical Sciences Polytechnic University of Marche Ancona Italy
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods Polytechnic University of Marche (Italy), Universidad Europea del Atlántico (Spain), and Jiangsu University (China) Ancona Italy
- International Joint Research, Laboratory of Intelligent Agriculture and Agri‐Products Processing Jiangsu University Zhenjiang China
| | - Wei Jia
- Department of Pharmacology and Pharmacy The University of Hong Kong Pokfulam Hong Kong SAR
| | - Qi Su
- Microbiota I‐Center Shatin Hong Kong SAR
- Department of Medicine and Therapeutics The Chinese University of Hong Kong Shatin Hong Kong SAR
| | - Xiaoqiang Ma
- Department of Food Science and Technology, School of Agriculture and Biology Shanghai Jiao Tong University Shanghai China
| | - Judith M. Rollinger
- Division of Pharmacognosy, Department of Pharmaceutical Sciences, Faculty of Life Sciences University of Vienna Vienna Austria
| | - Simon K.‐M. R. Rittmann
- Archaea Physiology & Biotechnology Group, Department of Functional and Evolutionary Ecology University of Vienna Vienna Austria
| | - Helen Sheridan
- The NatPro Centre & School of Pharmacy and Pharmaceutical Sciences Trinity College Dublin Dublin Ireland
- Université Bourgogne Europe/INSERM, 21000 Dijon and PHYNOHA Consulting Fontaine‐lès‐Dijon France
| | - John J. Walsh
- The NatPro Centre & School of Pharmacy and Pharmaceutical Sciences Trinity College Dublin Dublin Ireland
| | - Gérard Lizard
- Université Bourgogne Europe/INSERM, 21000 Dijon and PHYNOHA Consulting Fontaine‐lès‐Dijon France
| | - Tomasz M. Karpiński
- Department of Medical Microbiology Poznań University of Medical Sciences Poznań Poland
| | - Ana Sanches Silva
- University of Coimbra, Faculty of Pharmacy, Polo III, Azinhaga de Santa Comba Coimbra Portugal
- Centre for Animal Science Studies (CECA), ICETA University of Porto Porto Portugal
| | - Jakub Piwowarski
- Microbiota Lab, Department of Pharmaceutical Microbiology and Bioanalysis Medical University of Warsaw Warsaw Poland
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology Institute of Microbiology, Guangdong Academy of Sciences Guangzhou China
- School of Life & Health Sciences Fuyao University of Science & Technology Fuzhou Fujian China
| | - Tai‐Ping Fan
- School of Life & Health Sciences Fuyao University of Science & Technology Fuzhou Fujian China
| | - Francesca Giampieri
- Department of Clinical Sciences Polytechnic University of Marche Ancona Italy
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods Polytechnic University of Marche (Italy), Universidad Europea del Atlántico (Spain), and Jiangsu University (China) Ancona Italy
- Research Group on Food, Nutritional Biochemistry and Health Universidad Europea del Atlántico Santander Spain
- International Research Center for Food Nutrition and Safety Jiangsu University Zhenjiang China
| | - Adil El Midaoui
- Faculty of Sciences and Techniques Errachidia, Moulay Ismail University of Meknes Meknes Morocco
- Department of Pharmacology and Physiology, Faculty of Medicine University of Montreal Montreal Quebec Canada
| | - Ka‐Hing Wong
- Research Institute for Future Food The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
- Department of Food Science and Nutrition The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
| | - Ren‐You Gan
- Research Institute for Future Food The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
- Department of Food Science and Nutrition The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
| | - Ahmed Fatimi
- Chemical Science and Engineering Research Team (ERSIC), Department of Chemistry, Polydisciplinary Faculty of Beni Mellal (FPBM) Sultan Moulay Slimane University (USMS) Beni Mellal Morocco
| | - Atanas G. Atanasov
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
- Ludwig Boltzmann Institute Digital Health and Patient Safety Medical University of Vienna Vienna Austria
- Laboratory of Natural Products and Medicinal Chemistry (LNPMC), Center for Global Health Research, Saveetha Medical College and Hospital Saveetha Institute of Medical and Technical Sciences (SIMATS) Thandalam Chennai India
| |
Collapse
|
11
|
Wang J, Wang X, Jiang M, Lang T, Wan L, Dai J. 5-aminosalicylic acid alleviates colitis and protects intestinal barrier function by modulating gut microbiota in mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3681-3695. [PMID: 39352537 DOI: 10.1007/s00210-024-03485-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/23/2024] [Indexed: 04/10/2025]
Abstract
5-aminosalicylic acid (5-ASA) is widely used in the treatment of ulcerative colitis (UC), but its anti-inflammatory mechanism is complex and has not been fully understood. DSS model was used to test the effect of 5-ASA. Tight junction and Ki-67 were detected by western blot, immunofluorescence, and immunohistochemistry or qPCR. 16S rRNA gene sequencing of gut microbiota and subsequent bioinformatics and statistical analysis were performed to identify the specific bacteria which were associated with the treatment effect of 5-ASA. GC-MS was performed to test short-chain fatty acids (SCFAs). Antibiotic-treated mice were used to demonstrate the key role of endogenous gut microbiota. Here, we found that 5-ASA alleviated dextran sulfate sodium (DSS)-induced colitis in mice. Moreover, 5-ASA significantly repaired the intestinal barrier. At the molecular level, 5-ASA markedly raised the expression of tight junction proteins including JAM-A and occludin and cell proliferation marker Ki-67 in mice. In addition, bacterial 16S rRNA gene sequencing and bioinformatics analysis showed that 5-ASA significantly modulated the DSS-induced gut bacterial dysbiosis. In detail, it stimulated the growth of protective bacteria belonging to Faecalibaculum and Dubosiella, which were negatively correlated with colitis parameters, and blocked the expansion of pro-inflammatory bacteria such as Escherichia-Shigella and Oscillibacter, which were positively correlated with colitis in mice. Meanwhile, 5-ASA increased the cecal acetate level. Most notably, 5-ASA was no longer able to treat colitis and reverse gut barrier dysfunction in antibiotic-treated mice that lacked endogenous gut microbiota. Our data suggested that the anti-inflammatory activity of 5-ASA required the inherent intestinal flora, and the gut microbiota was a potential and effective target for the treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Jingjing Wang
- Shanghai Key Laboratory of Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxin Wang
- Shanghai Key Laboratory of Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingjie Jiang
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, P.R. China
| | - Tao Lang
- Shanghai Key Laboratory of Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Leilei Wan
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Juanjuan Dai
- Shanghai Key Laboratory of Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Intensive Care Unit, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, P.R. China.
| |
Collapse
|
12
|
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
|
13
|
Chen Y, Chen X, Lin S, Huang S, Li L, Hong M, Li J, Ma L, Ma J. Effects of psychological stress on inflammatory bowel disease via affecting the microbiota-gut-brain axis. Chin Med J (Engl) 2025; 138:664-677. [PMID: 39965932 PMCID: PMC11925421 DOI: 10.1097/cm9.0000000000003389] [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: 08/25/2024] [Indexed: 02/20/2025] Open
Abstract
ABSTRACT Inflammatory bowel disease (IBD) is an idiopathic intestinal inflammatory condition with chronic and relapsing manifestations and is characterized by a disturbance in the interplay between the intestinal microbiota, the gut, and the brain. The microbiota-gut-brain axis involves interactions among the nervous system, the neuroendocrine system, the gut microbiota, and the host immune system. Increasing published data indicate that psychological stress exacerbates the severity of IBD due to its negative effects on the microbiota-gut-brain axis, including alterations in the stress response of the hypothalamic-pituitary-adrenal (HPA) axis, the balance between the sympathetic nervous system and vagus nerves, the homeostasis of the intestinal flora and metabolites, and normal intestinal immunity and permeability. Although the current evidence is insufficient, psychotropic agents, psychotherapies, and interventions targeting the microbiota-gut-brain axis show the potential to improve symptoms and quality of life in IBD patients. Therefore, further studies that translate recent findings into therapeutic approaches that improve both physical and psychological well-being are needed.
Collapse
Affiliation(s)
- Yuhan Chen
- Shantou University Medical College, Shantou, Guangdong 515041, China
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Xiaofen Chen
- Shantou University Medical College, Shantou, Guangdong 515041, China
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Suqin Lin
- Medical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shengjun Huang
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Medical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lijuan Li
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Medical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Mingzhi Hong
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Medical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jianzhou Li
- Department of Diagnosis and Treatment Center of High Altitude Digestive Disease, The Second People’s Hospital of Xining, Xining, Qinghai 810003, China
| | - Lili Ma
- Department of Gastroenterology and Hepatology, Qinghai Provincial People’s Hospital, Xining, Qinghai 810007, China
| | - Juan Ma
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Department of Diagnosis and Treatment Center of High Altitude Digestive Disease, The Second People’s Hospital of Xining, Xining, Qinghai 810003, China
| |
Collapse
|
14
|
Chen T, Hong L, Wang P, Teng Q, Fang F, Liu Q. Protective Effect and Gut Microbiota Modulation of Grifola frondosa Antioxidant Peptides in Sodium Dextran Sulfate-Induced Ulcerative Colitis Mice. Biotechnol Appl Biochem 2025. [PMID: 39957377 DOI: 10.1002/bab.2734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/25/2025] [Indexed: 02/18/2025]
Abstract
Grifola frondosa antioxidant peptides (GFAP) were prepared through trypsin enzymolysis and characterized. This study conducted a comprehensive assessment of clinical symptoms, colon pathological injuries, levels of inflammatory factors, expression of inflammation-related proteins, and alterations in gut microbiota composition in mice with ulcerative colitis (UC). The findings demonstrated that GFAP effectively mitigated UC, alleviated mucosal damage, and reduced inflammatory infiltration. Specifically, GFAP administration resulted in significant reductions in pro-inflammatory cytokines IL-6, IL-1β, and TNF-α, while enhancing the expression levels of tight junction proteins such as Occludin and ZO-1. Additionally, GFAP treatment led to decreased levels of Toll-like receptor 4 (TLR-4), inducible nitric oxide synthase (iNOS), and TNF-α. Noteworthy, GFAP also influenced the gut microbiota by decreasing the abundance of Proteobacteria and increasing Bacteroidetes and Firmicutes. Moreover, specific bacteria like Bacteroides uniformis and Alistipes exhibited elevated abundances following GFAP treatment. In summary, GFAP exhibited preventive and protective effects against UC in mice by effectively alleviating clinical symptoms and modulating gut microbiota composition.
Collapse
Affiliation(s)
- Tong Chen
- Laboratory of Medicinal and Edible Fungi, Department of Vegetables, College of Horticulture, China Agricultural University, Beijing, China
| | - Linhai Hong
- Laboratory of Medicinal and Edible Fungi, Department of Vegetables, College of Horticulture, China Agricultural University, Beijing, China
| | - Peng Wang
- Laboratory of Medicinal and Edible Fungi, Department of Vegetables, College of Horticulture, China Agricultural University, Beijing, China
| | - Qian Teng
- Laboratory of Medicinal and Edible Fungi, Department of Vegetables, College of Horticulture, China Agricultural University, Beijing, China
| | - Fei Fang
- Laboratory of Medicinal and Edible Fungi, Department of Vegetables, College of Horticulture, China Agricultural University, Beijing, China
| | - Qinghong Liu
- Laboratory of Medicinal and Edible Fungi, Department of Vegetables, College of Horticulture, China Agricultural University, Beijing, China
| |
Collapse
|
15
|
Moutsoglou D, Syal A, Lopez S, Nelson EC, Chen L, Kabage AJ, Fischer M, Khoruts A, Vaughn BP, Staley C. Novel Microbial Engraftment Trajectories Following Microbiota Transplant Therapy in Ulcerative Colitis. J Crohns Colitis 2025; 19:jjae142. [PMID: 39240145 DOI: 10.1093/ecco-jcc/jjae142] [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: 05/28/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND AND AIMS Microbiota transplant therapy (MTT) is an emerging treatment for ulcerative colitis (UC). One proposed mechanism for the benefit of MTT is through engraftment of donor microbiota; however, engraftment kinetics are unknown. We identified SourceTracker as an efficient method both to determine engraftment and for the kinetic study of engrafting donor taxa to aid in determining the mechanism of how this therapy may treat UC. METHODS Ulcerative colitis patients received either encapsulated (drug name MTP-101C) or placebo capsules daily for 8 weeks followed by a 4-week washout period. Amplicon sequence data from donors and patients were analyzed using the Bayesian algorithm SourceTracker. RESULTS Twenty-seven patients were enrolled, 14 to placebo and 13 to MTT. Baseline Shannon and Chao1 indices negatively correlated with week 12 donor engraftment for patients treated with active drug capsules but not for placebo patients. SourceTracker engraftment positively correlated with the week 12 distance from donors measured using the Bray-Curtis similarity metric in treated patients but not with placebo. Engraftment at week 12 was significantly higher in the MTT group than in the placebo group. We identified engrafting taxa from donors in our patients and quantified the proportion of donor similarity or engraftment during weeks 1 through 8 (active treatment) and week 12, 4 weeks after the last dose. CONCLUSION SourceTracker can be used as a simple and reliable method to quantify donor microbial community engraftment and donor taxa contribution in patients with UC and other inflammatory conditions treated with MTT.
Collapse
Affiliation(s)
- Daphne Moutsoglou
- Department of Gastroenterology, Minneapolis VA Health Care System, MN 55417, USA
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Aneesh Syal
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Sharon Lopez
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Elizabeth C Nelson
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Lulu Chen
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Amanda J Kabage
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Monika Fischer
- Division of Gastroenterology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Byron P Vaughn
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Christopher Staley
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
16
|
Pribyl AL, Hugenholtz P, Cooper MA. A decade of advances in human gut microbiome-derived biotherapeutics. Nat Microbiol 2025; 10:301-312. [PMID: 39779879 DOI: 10.1038/s41564-024-01896-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025]
Abstract
Microbiome science has evolved rapidly in the past decade, with high-profile publications suggesting that the gut microbiome is a causal determinant of human health. This has led to the emergence of microbiome-focused biotechnology companies and pharmaceutical company investment in the research and development of gut-derived therapeutics. Despite the early promise of this field, the first generation of microbiome-derived therapeutics (faecal microbiota products) have only recently been approved for clinical use. Next-generation therapies based on readily culturable and as-yet-unculturable colonic bacterial species (with the latter estimated to comprise 63% of all detected species) have not yet progressed to pivotal phase 3 trials. This reflects the many challenges involved in developing a new class of drugs in an evolving field. Here we discuss the evolution of the live biotherapeutics field over the past decade, from the development of first-generation products to the emergence of rationally designed second- and third-generation live biotherapeutics. Finally, we present our outlook for the future of this field.
Collapse
Affiliation(s)
| | - Philip Hugenholtz
- The University of Queensland, School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, Brisbane, Queensland, Australia.
| | | |
Collapse
|
17
|
Marwaha K, Cain R, Asmis K, Czaplinski K, Holland N, Mayer DCG, Chacon J. Exploring the complex relationship between psychosocial stress and the gut microbiome: implications for inflammation and immune modulation. J Appl Physiol (1985) 2025; 138:518-535. [PMID: 39813028 DOI: 10.1152/japplphysiol.00652.2024] [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: 08/22/2024] [Revised: 12/16/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
There is growing interest in understanding the complex relationship between psychosocial stress and the human gastrointestinal microbiome (GIM). This review explores the potential physiological pathways connecting these two and how they contribute to a proinflammatory environment that can lead to the development and progression of the disease. Exposure to psychosocial stress triggers the activation of the sympathetic nervous system (SNS) and hypothalamic-pituitary axis (HPA), leading to various physiological responses essential for survival and coping with the stressor. However, chronic stress in susceptible individuals could cause sustained activation of HPA and SNS, leading to immune dysregulation consisting of redistribution of natural killer (NK) cells in the bloodstream, decreased function of T and B cells, and elevation of proinflammatory cytokines such as interleukin-1, interleukin-6, tumor necrotic factor-α, interferon-gamma. It also leads to disruption of the GIM composition and increased intestinal barrier permeability, contributing to GIM dysbiosis. The GIM dysbiosis and elevated cytokines can lead to reciprocal effects and further stimulate the HPA and SNS, creating a positive feedback loop that results in a proinflammatory state underlying the pathogenesis and progression of stress-associated cardiovascular, gastrointestinal, autoimmune, and psychiatric disorders. Understanding these relationships is critical for developing new strategies for managing stress-related health disorders.
Collapse
Affiliation(s)
- Komal Marwaha
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| | - Ryan Cain
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| | - Katherine Asmis
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| | - Katya Czaplinski
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| | - Nathan Holland
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| | - Darly C Ghislaine Mayer
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| | - Jessica Chacon
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| |
Collapse
|
18
|
Wu C, Lin B, Zhang H, Xu D, Gao R, Song R, Liu ZP, De Marinis Y. GCNPMDA: Human microbe-disease association prediction by hierarchical graph convolutional network with layer attention. Biomed Signal Process Control 2025; 100:107004. [DOI: 10.1016/j.bspc.2024.107004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
19
|
Qiao Y, He C, Xia Y, Ocansey DKW, Mao F. Intestinal mucus barrier: A potential therapeutic target for IBD. Autoimmun Rev 2025; 24:103717. [PMID: 39662652 DOI: 10.1016/j.autrev.2024.103717] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Intestinal mucus, a viscoelastic medium with mucin2 (MUC2) as its main component, covers the surface of intestinal epithelial cells and protects the intestine from invasion, forming the first barrier of the intestinal tract. Unlike the small intestine, where the mucus layer is a single layer, the colonic mucus layer can be divided into a sterile inner layer and an outer layer with bacterial colonization. Many of the substances in the mucus layer have beneficial effects on the intestinal epithelium, but the mucus layer is often affected by a variety of factors, mainly microbiological, dietary, and immunological. Inflammatory bowel disease (IBD) is a disease of increasing morbidity worldwide, with a complex etiology and a high relapse rate. In recent years, the mucus barrier in IBD has received increasing attention and is considered a key factor in the pathogenesis of IBD. Loss of goblet cells (GCs) and changes in the composition and properties of the mucus layer material are commonly found in the colon of IBD patients. Damage to the mucus layer may make it easier for microorganisms to access the intestinal epithelium and cause inflammation. There are currently a number of herbs and other therapies that can be used to treat IBD and repair the damaged mucus barrier. This review highlights the important role of the mucus layer in IBD and the therapies that target the mucus layer in IBD.
Collapse
Affiliation(s)
- Yaru Qiao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Changer He
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang 212399, Jiangsu, PR China
| | - Yuxuan Xia
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China; Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast CC0959347, Ghana
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
20
|
Shang J, Del Valle DM, Britton GJ, Mead K, Rajpal U, Chen-Liaw A, Mogno I, Li Z, Menon R, Gonzalez-Kozlova E, Elkrief A, Peled JU, Gonsalves TR, Shah NJ, Postow M, Colombel JF, Gnjatic S, Faleck DM, Faith JJ. Baseline colitogenicity and acute perturbations of gut microbiota in immunotherapy-related colitis. J Exp Med 2025; 222:e20232079. [PMID: 39666007 PMCID: PMC11636624 DOI: 10.1084/jem.20232079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 09/17/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024] Open
Abstract
Immunotherapy-related colitis (irC) frequently emerges as an immune-related adverse event during immune checkpoint inhibitor therapy and is presumably influenced by the gut microbiota. We longitudinally studied microbiomes from 38 ICI-treated cancer patients. We compared 13 ICI-treated subjects who developed irC against 25 ICI-treated subjects who remained irC-free, along with a validation cohort. Leveraging a preclinical mouse model, predisease stools from irC subjects induced greater colitigenicity upon transfer to mice. The microbiota during the first 10 days of irC closely resembled inflammatory bowel disease microbiomes, with reduced diversity, increased Proteobacteria and Veillonella, and decreased Faecalibacterium, which normalized before irC remission. These findings highlight the irC gut microbiota as functionally distinct but phylogenetically similar to non-irC and healthy microbiomes, with the exception of an acute, transient disruption early in irC. We underscore the significance of longitudinal microbiome profiling in developing clinical avenues to detect, monitor, and mitigate irC in ICI therapy cancer patients.
Collapse
Affiliation(s)
- Joan Shang
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diane Marie Del Valle
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Graham J. Britton
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - K.R. Mead
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Urvija Rajpal
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alice Chen-Liaw
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ilaria Mogno
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhihua Li
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Edgar Gonzalez-Kozlova
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arielle Elkrief
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan U. Peled
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Tina Ruth Gonsalves
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Neil J. Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Michael Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David M. Faleck
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Jeremiah J. Faith
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
21
|
Kim J, Gunathilake M, Yeo HY, Oh JH, Kim BC, Han N, Kim B, Pyun H, Lim MY, Nam YD, Chang HJ. Fecal Microbial Dysbiosis Is Associated with Colorectal Cancer Risk in a Korean Population. Cancer Res Treat 2025; 57:198-211. [PMID: 39054623 PMCID: PMC11729318 DOI: 10.4143/crt.2024.382] [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: 04/18/2024] [Accepted: 07/25/2024] [Indexed: 07/27/2024] Open
Abstract
PURPOSE The association between the fecal microbiota and colorectal cancer (CRC) risk has been suggested in epidemiologic studies. However, data from large-scale population-based studies are lacking. MATERIALS AND METHODS In this case-control study, we recruited 283 CRC patients from the Center for Colorectal Cancer, National Cancer Center Hospital, Korea to perform 16S rRNA gene sequencing of fecal samples. A total of 283 age- and sex-matched healthy participants were selected from 890 cohort of healthy Koreans that are publicly available (PRJEB33905). The microbial dysbiosis index (MDI) was calculated based on the differentially abundant species. The association between MDI and CRC risk was observed using conditional logistic regression. Sparse Canonical Correlation Analysis was performed to integrate species data with microbial pathways obtained by PICRUSt2. RESULTS There is a significant divergence of the microbial composition between CRC patients and controls (permutational multivariate analysis of variance p=0.001). Those who were in third tertile of the MDI showed a significantly increased risk of CRC in the total population (odds ratio [OR], 6.93; 95% confidence interval [CI], 3.98 to 12.06; p-trend < 0.001) compared to those in the lowest tertile. Similar results were found for men (OR, 6.28; 95% CI, 3.04 to 12.98; p-trend < 0.001) and women (OR, 7.39; 95% CI, 3.10 to 17.63; p-trend < 0.001). Bacteroides coprocola and Bacteroides plebeius species and 12 metabolic pathways were interrelated in healthy controls that explain 91% covariation across samples. CONCLUSION Dysbiosis in the fecal microbiota may be associated with an increased risk of CRC. Due to the potentially modifiable nature of the gut microbiota, our findings may have implications for CRC prevention among Koreans.
Collapse
Affiliation(s)
- Jeongseon Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Madhawa Gunathilake
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Hyun Yang Yeo
- Department of Cancer Diagnostics, Research Institute, National Cancer Center, Goyang, Korea
| | - Jae Hwan Oh
- Center for Colorectal Cancer, National Cancer Center Hospital, National Cancer Center, Goyang, Korea
| | - Byung Chang Kim
- Center for Colorectal Cancer, National Cancer Center Hospital, National Cancer Center, Goyang, Korea
| | - Nayoung Han
- Department of Pathology, National Cancer Center Hospital, National Cancer Center, Goyang, Korea
| | - Bun Kim
- Department of Cancer Diagnostics, Research Institute, National Cancer Center, Goyang, Korea
- Center for Colorectal Cancer, National Cancer Center Hospital, National Cancer Center, Goyang, Korea
| | - Hyojin Pyun
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Mi Young Lim
- Personalized Diet Research Group, Food Functionality Research Division, Korea Food Research Institute, Wanju, Korea
| | - Young-Do Nam
- Personalized Diet Research Group, Food Functionality Research Division, Korea Food Research Institute, Wanju, Korea
| | - Hee Jin Chang
- Department of Cancer Diagnostics, Research Institute, National Cancer Center, Goyang, Korea
- Center for Colorectal Cancer, National Cancer Center Hospital, National Cancer Center, Goyang, Korea
- Department of Pathology, National Cancer Center Hospital, National Cancer Center, Goyang, Korea
| |
Collapse
|
22
|
Gamada H, Funayama T, Nakagawa T, Sunami T, Sakashita K, Inomata K, Okuwaki S, Asada T, Shibao Y, Miura K, Noguchi H, Takahashi H, Shiina I, Nakagawa T, Yamazaki M, Koda M. Comparison of Disc Degeneration between Pyogenic Spondylitis and Noninfected Lumbar Spondylosis: A Multicenter Retrospective Study with Propensity Score Matching. Spine Surg Relat Res 2024; 8:616-622. [PMID: 39659380 PMCID: PMC11625711 DOI: 10.22603/ssrr.2024-0032] [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: 02/09/2024] [Accepted: 03/15/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction Disc degeneration is a risk factor of pyogenic spondylitis. However, its degree in patients with pyogenic spondylitis is unknown. This study aimed to determine differences in disc degeneration between patients with pyogenic spondylitis and those with noninfectious lumbar spondylosis. Methods A total of 85 patients with lumbar pyogenic spondylitis (the infected group) and 156 with lumbar spondylosis who underwent posterior lumbar interbody fusion (the noninfected group) were retrospectively evaluated. Patients with a previous history of spinal fusion, tuberculous spondylitis, and multilevel infection and those receiving dialysis were excluded. Magnetic resonance imaging of the lumbar spine was conducted. Each disc at the L1/2-L5/S levels was graded. The total score of the four discs, excluding the affected disc, was used as the modified disc degenerative disease (DDD) score. Propensity score matching was performed using independent variables such as age, sex, diabetes mellitus, cancer, and steroid use. The modified DDD scores at all and each disc level were compared between the two matched groups. Results After matching, 48 patients in the infected group and 88 in the noninfected group were finally included in the study. The mean modified DDD scores of the infected and noninfected groups were 7.63 and 5.40, respectively. The modified DDD scores at all and each disc level were higher in the infected group than in the noninfected group. Conclusions The incidence of disc degeneration at all and each disc level was higher in patients with pyogenic spondylitis than in those with noninfectious lumbar spondylosis.
Collapse
Affiliation(s)
- Hisanori Gamada
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Orthopaedic Surgery, Ibaraki Western Medical Center, Chikusei, Japan
| | - Toru Funayama
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Orthopaedic Surgery, Kenpoku Medical Center Takahagi Kyodo Hospital, Takahagi, Japan
| | - Takane Nakagawa
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Orthopaedic Surgery, Moriya Daiichi General Hospital, Moriya, Japan
| | - Takahiro Sunami
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kotaro Sakashita
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kento Inomata
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shun Okuwaki
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tomoyuki Asada
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yosuke Shibao
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Orthopaedic Surgery, Ibaraki Western Medical Center, Chikusei, Japan
| | - Kousei Miura
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroshi Noguchi
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroshi Takahashi
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Itsuo Shiina
- Department of Orthopaedic Surgery, Moriya Daiichi General Hospital, Moriya, Japan
| | - Tsukasa Nakagawa
- Department of Orthopaedic Surgery, Ibaraki Western Medical Center, Chikusei, Japan
| | - Masashi Yamazaki
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masao Koda
- Department of Orthopaedic Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
23
|
Mallick R, Basak S, Das RK, Banerjee A, Paul S, Pathak S, Duttaroy AK. Roles of the gut microbiota in human neurodevelopment and adult brain disorders. Front Neurosci 2024; 18:1446700. [PMID: 39659882 PMCID: PMC11628544 DOI: 10.3389/fnins.2024.1446700] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024] Open
Abstract
Growing evidence demonstrates the connection between gut microbiota, neurodevelopment, and adult brain function. Microbial colonization occurs before the maturation of neural systems and its association with brain development. The early microbiome interactions with the gut-brain axis evolved to stimulate cognitive activities. Gut dysbiosis can lead to impaired brain development, growth, and function. Docosahexaenoic acid (DHA) is critically required for brain structure and function, modulates gut microbiota, and impacts brain activity. This review explores how gut microbiota influences early brain development and adult functions, encompassing the modulation of neurotransmitter activity, neuroinflammation, and blood-brain barrier integrity. In addition, it highlights processes of how the gut microbiome affects fetal neurodevelopment and discusses adult brain disorders.
Collapse
Affiliation(s)
- Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Ranjit K. Das
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Queretaro, Mexico
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| | - Asim K. Duttaroy
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
24
|
Seo SM, Kim NW, Yoo ES, Lee JH, Kang AR, Jeong HB, Shim WY, Kim DH, Park YJ, Bae K, Yoon KA, Choi YK. Development of a novel complex inflammatory bowel disease mouse model: Reproducing human inflammatory bowel disease etiologies in mice. PLoS One 2024; 19:e0311310. [PMID: 39570897 PMCID: PMC11581264 DOI: 10.1371/journal.pone.0311310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/30/2024] [Indexed: 11/24/2024] Open
Abstract
Inflammatory bowel disease (IBD), caused by environmental factors associated with the host's genetic traits, is represented by Crohn's disease and ulcerative colitis. Despite the increasing number of patients with IBD, the current treatment is limited to symptomatic therapy. A complex IBD model mimicking the human IBD etiology is required to overcome this limitation. Herein, we developed novel complex IBD models using interleukin 2 receptor subunit gamma (Il2rg)-deficient mice, high-fat diet, dextran sodium sulfate, and Citrobacter rodentium. The more IBD factors applied complexly, colon length shortened and inflammation worsened. The levels of pro-inflammatory cytokines increased with increased IBD factors. Anti-inflammatory cytokine decreased in all factors application but increased in Il2rg deficiency and Westernized diet combination. Additionally, the pro-inflammatory transcription factors and leaky intestinal epithelial marker were upregulated by a combination of IBD factors. Species diversity decreased with IBD factors. Phylogenetic diversity decreased as IBD factors were applied but increased with combined Il2rg deficiency and Westernized diet. The more IBD factors applied complexly, the more severe the dysbiosis. Finally, we developed a novel complex IBD model using various IBD factors. This model more closely mimic human IBD based on colonic inflammation and dysbiosis than the previous models. Based on these results, our novel complex IBD model could be a valuable tool for further IBD research.
Collapse
Affiliation(s)
- Sun-Min Seo
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Na-Won Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Eun-Seon Yoo
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Ji-Hun Lee
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Ah-Reum Kang
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Han-Bi Jeong
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Won-Yong Shim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Dong-Hyun Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Young-Jun Park
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Kieun Bae
- Department of Veterinary Biochemistry, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Kyong-Ah Yoon
- Department of Veterinary Biochemistry, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
25
|
Boldyreva LV, Evtushenko AA, Lvova MN, Morozova KN, Kiseleva EV. Underneath the Gut-Brain Axis in IBD-Evidence of the Non-Obvious. Int J Mol Sci 2024; 25:12125. [PMID: 39596193 PMCID: PMC11594934 DOI: 10.3390/ijms252212125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
The gut-brain axis (GBA) plays a pivotal role in human health and wellness by orchestrating complex bidirectional regulation and influencing numerous critical processes within the body. Over the past decade, research has increasingly focused on the GBA in the context of inflammatory bowel disease (IBD). Beyond its well-documented effects on the GBA-enteric nervous system and vagus nerve dysregulation, and gut microbiota misbalance-IBD also leads to impairments in the metabolic and cellular functions: metabolic dysregulation, mitochondrial dysfunction, cationic transport, and cytoskeleton dysregulation. These systemic effects are currently underexplored in relation to the GBA; however, they are crucial for the nervous system cells' functioning. This review summarizes the studies on the particular mechanisms of metabolic dysregulation, mitochondrial dysfunction, cationic transport, and cytoskeleton impairments in IBD. Understanding the involvement of these processes in the GBA may help find new therapeutic targets and develop systemic approaches to improve the quality of life in IBD patients.
Collapse
Affiliation(s)
- Lidiya V. Boldyreva
- Scientific-Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia;
| | - Anna A. Evtushenko
- Scientific-Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia;
| | - Maria N. Lvova
- Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.N.L.); (K.N.M.); (E.V.K.)
| | - Ksenia N. Morozova
- Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.N.L.); (K.N.M.); (E.V.K.)
| | - Elena V. Kiseleva
- Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.N.L.); (K.N.M.); (E.V.K.)
| |
Collapse
|
26
|
Kao D, Wong K, Jijon H, Moayyedi P, Franz R, McDougall C, Hotte N, Panaccione R, Semlacher E, Kroeker KI, Peerani F, MacDonald KV, Xu H, Narula N, Turbide C, Marshall DA, Madsen KL. Preliminary Results From a Multicenter, Randomized Trial Using Fecal Microbiota Transplantation to Induce Remission in Patients With Mild-to-Moderate Crohn's Disease. Am J Gastroenterol 2024; 120:00000434-990000000-01438. [PMID: 39530534 PMCID: PMC12124204 DOI: 10.14309/ajg.0000000000003196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Fecal microbiota transplantation (FMT) has shown promise at inducing remission in ulcerative colitis. This study is the first of its kind to evaluate the efficacy and safety of FMT at inducing remission in Crohn's disease (CD). METHODS This double-blind, placebo-controlled trial was conducted in 3 Canadian academic centers; randomized patients with mild-to-moderate CD received FMT or placebo. The first treatment was administered by colonoscopy followed by weekly oral capsules for 7 weeks. Primary end point was clinical and endoscopic remission at week 8. Secondary outcomes included clinical and endoscopic response, adverse events, and health-related quality of life using generic and disease-specific instruments. RESULTS From July 2017 to June 2021, 21 and 13 patients were randomized to FMT and placebo groups, respectively. The trial terminated early due to futility. At week 8, 0% (0/15) of patients in the FMT group versus 8.3% (1/11) in the placebo group reached the primary end point of combined clinical and endoscopic remission as per protocol analysis. There were no differences between the groups in clinical or endoscopic responses. One patient in each group had worsening of CD. Although both groups experienced statistically significant improvements in health-related quality of life, only the FMT group had a significant decrease in activity impairment. Although there were no significant changes in microbial diversity or composition, patients who achieved clinical response became more similar to their donors in stool microbial composition. DISCUSSION FMT was not effective at inducing clinical and endoscopic remission in CD using the FMT regimen in this study. Future studies may use other strategies to enhance treatment response, including longer intervention, antibiotic pretreatment, optimized donor-recipient pairing, and concomitant anti-inflammatory diet, and biologic or small molecule therapies.
Collapse
Affiliation(s)
- Dina Kao
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Karen Wong
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Humberto Jijon
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Calgary, Calgary, Alberta, Canada
| | - Paul Moayyedi
- Department of Medicine, Division of Gastroenterology, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Rose Franz
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Chelsea McDougall
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Naomi Hotte
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Remo Panaccione
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Calgary, Calgary, Alberta, Canada
| | - Eric Semlacher
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Karen I. Kroeker
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Farhad Peerani
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Karen V. MacDonald
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Huiping Xu
- Biostatstics & Health Data Sciences, School of Public Health, Indiana University, Indianapolis, Indiana, USA
| | - Neeraj Narula
- Department of Medicine, Division of Gastroenterology, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Christian Turbide
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Calgary, Calgary, Alberta, Canada
| | - Deborah A. Marshall
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Karen L. Madsen
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
27
|
Wang L, Zheng W, Men Q, Ren X, Song S, Ai C. Curcumin-loaded polysaccharide microparticles alleviated DSS-induced ulcerative colitis by improving intestinal microecology and regulating MAPK/NF-κB/Nrf2/NLRP3 pathways. Int J Biol Macromol 2024; 281:136687. [PMID: 39427805 DOI: 10.1016/j.ijbiomac.2024.136687] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/19/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Curcumin (Cur) exerts many benefits on the host, but its application is limited by its poor bioavailability. In this study, composite polysaccharide microparticles loading Cur (Cur-CPM) was prepared by food-grade materials and gel technology. Its properties were analyzed via the in vitro and in vivo models, and then its benefit on gut health was assessed in DSS-treated mice. Compared to free Cur, CPM extended the residence time and absorption efficiency of Cur in the intestine, effectively ameliorating the symptoms of colitis. Cur-CPM alleviated colonic inflammation by inhibiting the activation of the MAPK and NF-κB pathways and suppressing NLRP3 inflammasome activity, affecting the expression of inflammation-related cytokines and mediators. In addition, Cur-CPM regulated the levels of antioxidants and oxidants in the colon tissues via Nrf2 activation, alleviating oxidative stress. Cur-CPM protected gut barrier function by maintaining the integrity of colonic mucosal layer and tight junction. The underlying mechanism can be attributed not only to the anti-inflammatory and antioxidant activities of Cur but also to modulation of Cur and CPM on the gut microbiota and metabolites. It suggests that Cur-CPM holds the potential to be developed as a functional component to enhance gut health.
Collapse
Affiliation(s)
- Lu Wang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Weiyun Zheng
- School of Agronomy and Life Science, Shanxi Datong University, Datong 037009, PR China
| | - Qiuyue Men
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Xiaomeng Ren
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuang Song
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunqing Ai
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
28
|
Huang Y, Wang Y, Huang X, Yu X. Unveiling the overlooked fungi: the vital of gut fungi in inflammatory bowel disease and colorectal cancer. Gut Pathog 2024; 16:59. [PMID: 39407244 PMCID: PMC11481806 DOI: 10.1186/s13099-024-00651-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
The fungi of the human microbiota play important roles in the nutritional metabolism and immunological balance of the host. Recently, research has increasingly emphasised the role of fungi in modulating inflammation in intestinal diseases and maintaining health in this environment. It is therefore necessary to understand more clearly the interactions and mechanisms of the microbiota/pathogen/host relationship and the resulting inflammatory processes, as well as to offer new insights into the prevention, diagnosis and treatment of inflammatory bowel disease (IBD), colorectal cancer (CRC) and other intestinal pathologies. In this review, we comprehensively elucidate the fungal-associated pathogenic mechanisms of intestinal inflammation in IBD and related CRC, with an emphasis on three main aspects: the direct effects of fungi and their metabolites on the host, the indirect effects mediated by interactions with other intestinal microorganisms and the immune regulation of the host. Understanding these mechanisms will enable the development of innovative approaches based on the use of fungi from the resident human microbiota such as dietary interventions, fungal probiotics and faecal microbiota transplantation in the prevention, diagnosis and treatment of intestinal diseases.
Collapse
Affiliation(s)
- Yilin Huang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Yang Wang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaotian Huang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Xiaomin Yu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
29
|
Casali L, Stella GM. The Microbiota in Children and Adolescents with Asthma. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1175. [PMID: 39457140 PMCID: PMC11505771 DOI: 10.3390/children11101175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/20/2024] [Accepted: 09/09/2024] [Indexed: 10/28/2024]
Abstract
The role of the respiratory microbiome has been deeply explored for at least two decades. Its characterization using modern methods is now well-defined, and the impacts of many microorganisms on health and diseases have been elucidated. Moreover, the acquired knowledge in related fields enables patient stratification based on their risk for disease onset, and the microbiome can play a role in defining possible phenotypes. The interplay between the lung and gut microbiomes is crucial in determining the microbial composition and immuno-inflammatory reaction. Asthma is still not a well-defined condition, where hyperreactivity and the immune system play important roles. In this disease, the microbiome is mostly represented by Proteobacteria, Streptococcus, and Veillonella, while Cytomegalovirus and Epstein-Barr viruses are the most prevalent viruses. A mycobiome may also be present. The passage from infancy to adolescence is examined by evaluating both the clinical picture and its relationship with possible variations of the microbiome and its effects on asthma. Otherwise, asthma is considered a heterogeneous disease that often starts in childhood and follows a particular personalized track, where adolescence plays a pivotal role in future prognosis. Under this point of view, the microbiota, with its possible variations due to many factors, both internal and external, can modify its composition; consequently, its inflammatory action and role in the immunological response has obvious consequences on the clinical conditions.
Collapse
Affiliation(s)
- Lucio Casali
- Unit of Respiratory Diseases, University of Perugia, 06121 Perugia, Italy;
| | - Giulia Maria Stella
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy
- Unit of Respiratory Diseases, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
30
|
Toto F, Marangelo C, Scanu M, De Angelis P, Isoldi S, Abreu MT, Cucchiara S, Stronati L, Del Chierico F, Putignani L. A Novel Microbial Dysbiosis Index and Intestinal Microbiota-Associated Markers as Tools of Precision Medicine in Inflammatory Bowel Disease Paediatric Patients. Int J Mol Sci 2024; 25:9618. [PMID: 39273567 PMCID: PMC11395508 DOI: 10.3390/ijms25179618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Recent evidence indicates that the gut microbiota (GM) has a significant impact on the inflammatory bowel disease (IBD) progression. Our aim was to investigate the GM profiles, the Microbial Dysbiosis Index (MDI) and the intestinal microbiota-associated markers in relation to IBD clinical characteristics and disease state. We performed 16S rRNA metataxonomy on both stools and ileal biopsies, metabolic dysbiosis tests on urine and intestinal permeability and mucosal immunity activation tests on the stools of 35 IBD paediatric patients. On the GM profile, we assigned the MDI to each patient. In the statistical analyses, the MDI was correlated with clinical parameters and intestinal microbial-associated markers. In IBD patients with high MDI, Gemellaceae and Enterobacteriaceae were increased in stools, and Fusobacterium, Haemophilus and Veillonella were increased in ileal biopsies. Ruminococcaceae and WAL_1855D were enriched in active disease condition; the last one was also positively correlated to MDI. Furthermore, the MDI results correlated with PUCAI and Matts scores in ulcerative colitis patients (UC). Finally, in our patients, we detected metabolic dysbiosis, intestinal permeability and mucosal immunity activation. In conclusion, the MDI showed a strong association with both severity and activity of IBD and a positive correlation with clinical scores, especially in UC. Thus, this evidence could be a useful tool for the diagnosis and prognosis of IBD.
Collapse
Affiliation(s)
- Francesca Toto
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Chiara Marangelo
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Matteo Scanu
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Paola De Angelis
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Sara Isoldi
- Pediatric Gastroenterology and Hepatology Unit, Santobono-Pausilipon Children's Hospital, 80122 Naples, Italy
| | - Maria Teresa Abreu
- Crohn's and Colitis Center, Division of Digestive Health and Liver Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Salvatore Cucchiara
- Maternal Child Health Department, Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, 00185 Rome, Italy
| | - Laura Stronati
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Federica Del Chierico
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| |
Collapse
|
31
|
Sardar P, Almeida A, Pedicord VA. Integrating functional metagenomics to decipher microbiome-immune interactions. Immunol Cell Biol 2024; 102:680-691. [PMID: 38952337 DOI: 10.1111/imcb.12798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 07/03/2024]
Abstract
Microbial metabolites can be viewed as the cytokines of the microbiome, transmitting information about the microbial and metabolic environment of the gut to orchestrate and modulate local and systemic immune responses. Still, many immunology studies focus solely on the taxonomy and community structure of the gut microbiota rather than its functions. Early sequencing-based microbiota profiling approaches relied on PCR amplification of small regions of bacterial and fungal genomes to facilitate identification of the microbes present. However, recent microbiome analysis methods, particularly shotgun metagenomic sequencing, now enable culture-independent profiling of microbiome functions and metabolites in addition to taxonomic characterization. In this review, we showcase recent advances in functional metagenomics methods and applications and discuss the current limitations and potential avenues for future development. Importantly, we highlight a few examples of key areas of opportunity in immunology research where integrating functional metagenomic analyses of the microbiome can substantially enhance a mechanistic understanding of microbiome-immune interactions and their contributions to health and disease states.
Collapse
Affiliation(s)
- Puspendu Sardar
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Alexandre Almeida
- Department of Veterinary Medicine, University of Cambridge School of Biological Sciences, Cambridge, UK
| | - Virginia A Pedicord
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
32
|
Zhang L, Lu J. Rosemary (Rosmarinus officinalis L.) polyphenols and inflammatory bowel diseases: Major phytochemicals, functional properties, and health effects. Fitoterapia 2024; 177:106074. [PMID: 38906386 DOI: 10.1016/j.fitote.2024.106074] [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: 02/25/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
Major polyphenols in Rosmarinus officinalis L. primarily consist of phenolic acids, phenolic diterpenes, and flavonoids, all of which have pharmacological properties including anti-inflammatory and antibacterial characteristics. Numerous in vitro and animal studies have found that rosemary polyphenols have the potential to decrease the severity of intestinal inflammation. The beneficial effects of rosemary polyphenols were associated with anti-inflammatory properties, including improved gut barrier (increased mucus secretion and tight junction), increased antioxidant enzymes, inhibiting inflammatory pathways and cytokines (downregulation of NF-κB, NLRP3 inflammasomes, STAT3 and activation of Nrf2), and modulating gut microbiota community (increased core probiotics and SCFA-producing bacteria, and decreased potential pathogens) and metabolism (changes in SCFA and bile acid metabolites). This paper provides a better understanding of the anti-inflammatory properties of rosemary polyphenols and suggests that rosemary polyphenols might be employed as strong anti-inflammatory agents to prevent intestinal inflammation and lower the risk of inflammatory bowel disease and related diseases.
Collapse
Affiliation(s)
- Lianhua Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Jie Lu
- China Animal Husbandry Group, Beijing 100070, China
| |
Collapse
|
33
|
Lin Q, Dorsett Y, Mirza A, Tremlett H, Piccio L, Longbrake EE, Choileain SN, Hafler DA, Cox LM, Weiner HL, Yamamura T, Chen K, Wu Y, Zhou Y. Meta-analysis identifies common gut microbiota associated with multiple sclerosis. Genome Med 2024; 16:94. [PMID: 39085949 PMCID: PMC11293023 DOI: 10.1186/s13073-024-01364-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/12/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Previous studies have identified a diverse group of microbial taxa that differ between patients with multiple sclerosis (MS) and the healthy population. However, interpreting findings on MS-associated microbiota is challenging, as there is no true consensus. It is unclear whether there is gut microbiota commonly altered in MS across studies. METHODS To answer this, we performed a meta-analysis based on the 16S rRNA gene sequencing data from seven geographically and technically diverse studies comprising a total of 524 adult subjects (257 MS and 267 healthy controls). Analysis was conducted for each individual study after reprocessing the data and also by combining all data together. The blocked Wilcoxon rank-sum test and linear mixed-effects regression were used to identify differences in microbial composition and diversity between MS and healthy controls. Network analysis was conducted to identify bacterial correlations. A leave-one-out sensitivity analysis was performed to ensure the robustness of the findings. RESULTS The microbiome community structure was significantly different between studies. Re-analysis of data from individual studies revealed a lower relative abundance of Prevotella in MS across studies, compared to controls. Meta-analysis found that although alpha and beta diversity did not differ between MS and controls, a higher abundance of Actinomyces and a lower abundance of Faecalibacterium were reproducibly associated with MS. Additionally, network analysis revealed that the recognized negative Bacteroides-Prevotella correlation in controls was disrupted in patients with MS. CONCLUSIONS Our meta-analysis identified common gut microbiota associated with MS across geographically and technically diverse studies.
Collapse
Affiliation(s)
- Qingqi Lin
- Department of Computer Science and Engineering, University of Connecticut, Storrs, CT, USA
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Yair Dorsett
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Ali Mirza
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Helen Tremlett
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Laura Piccio
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Brain and Mind Centre, School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Erin E Longbrake
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Siobhan Ni Choileain
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - David A Hafler
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Laura M Cox
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham & Women's Hospital, Boston, MA, 02115, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham & Women's Hospital, Boston, MA, 02115, USA
| | - Takashi Yamamura
- Department of Immunology, National Institute of Neuroscience, Tokyo, Japan
| | - Kun Chen
- Department of Statistics, University of Connecticut, Storrs, CT, USA
| | - Yufeng Wu
- Department of Computer Science and Engineering, University of Connecticut, Storrs, CT, USA
| | - Yanjiao Zhou
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
34
|
Chen W, Li Y, Wang W, Gao S, Hu J, Xiang B, Wu D, Jiao N, Xu T, Zhi M, Zhu L, Zhu R. Enhanced microbiota profiling in patients with quiescent Crohn's disease through comparison with paired healthy first-degree relatives. Cell Rep Med 2024; 5:101624. [PMID: 38942021 PMCID: PMC11293350 DOI: 10.1016/j.xcrm.2024.101624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 04/09/2024] [Accepted: 06/07/2024] [Indexed: 06/30/2024]
Abstract
Prior studies indicate no correlation between the gut microbes of healthy first-degree relatives (HFDRs) of patients with Crohn's disease (CD) and the development of CD. Here, we utilize HFDRs as controls to examine the microbiota and metabolome in individuals with active (CD-A) and quiescent (CD-R) CD, thereby minimizing the influence of genetic and environmental factors. When compared to non-relative controls, the use of HFDR controls identifies fewer differential taxa. Faecalibacterium, Dorea, and Fusicatenibacter are decreased in CD-R, independent of inflammation, and correlated with fecal short-chain fatty acids (SCFAs). Validation with a large multi-center cohort confirms decreased Faecalibacterium and other SCFA-producing genera in CD-R. Classification models based on these genera distinguish CD from healthy individuals and demonstrate superior diagnostic power than models constructed with markers identified using unrelated controls. Furthermore, these markers exhibited limited discriminatory capabilities for other diseases. Finally, our results are validated across multiple cohorts, underscoring their robustness and potential for diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Wanning Chen
- Department of Gastroenterology, the Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200072, P.R. China
| | - Yichen Li
- Medical College, Jiaying University, Meizhou 514031, P. R. China; Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, P.R. China; Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, P.R. China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology; Biomedical Innovation Center; The Sixth Affiliated Hospital, Sun Yat-sen University; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou 510655, P.R. China
| | - Wenxia Wang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, P.R. China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology; Biomedical Innovation Center; The Sixth Affiliated Hospital, Sun Yat-sen University; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou 510655, P.R. China
| | - Sheng Gao
- Department of Gastroenterology, the Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200072, P.R. China
| | - Jun Hu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology; Biomedical Innovation Center; The Sixth Affiliated Hospital, Sun Yat-sen University; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou 510655, P.R. China; Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, P.R. China
| | - Bingjie Xiang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology; Biomedical Innovation Center; The Sixth Affiliated Hospital, Sun Yat-sen University; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou 510655, P.R. China; Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, P.R. China
| | - Dingfeng Wu
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310058, Zhejiang, P.R. China
| | - Na Jiao
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310058, Zhejiang, P.R. China
| | - Tao Xu
- Medical College, Jiaying University, Meizhou 514031, P. R. China; Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, P.R. China
| | - Min Zhi
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology; Biomedical Innovation Center; The Sixth Affiliated Hospital, Sun Yat-sen University; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou 510655, P.R. China; Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, P.R. China.
| | - Lixin Zhu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, P.R. China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology; Biomedical Innovation Center; The Sixth Affiliated Hospital, Sun Yat-sen University; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou 510655, P.R. China.
| | - Ruixin Zhu
- Department of Gastroenterology, the Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200072, P.R. China.
| |
Collapse
|
35
|
Koester ST, Chow A, Pepper-Tunick E, Lee P, Eckert M, Brenchley L, Gardner P, Song HJ, Li N, Schiffenbauer A, Volochayev R, Bayat N, McLean JS, Rider LG, Shenoi S, Stevens AM, Dey N. Familial clustering of dysbiotic oral and fecal microbiomes in juvenile dermatomyositis. Sci Rep 2024; 14:16158. [PMID: 38997299 PMCID: PMC11245510 DOI: 10.1038/s41598-024-60225-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/19/2024] [Indexed: 07/14/2024] Open
Abstract
Juvenile dermatomyositis (JDM) is a rare immune-mediated disease of childhood with putative links to microbial exposures. In this multi-center, prospective, observational cohort study, we evaluated whether JDM is associated with discrete oral and gut microbiome signatures. We generated 16S rRNA sequencing data from fecal, saliva, supragingival, and subgingival plaque samples from JDM probands (n = 28). To control for genetic and environmental determinants of microbiome community structure, we also profiled microbiomes of unaffected family members (n = 27 siblings, n = 26 mothers, and n = 17 fathers). Sample type (oral-vs-fecal) and nuclear family unit were the predominant variables explaining variance in microbiome diversity, more so than having a diagnosis of JDM. The oral and gut microbiomes of JDM probands were more similar to their own unaffected siblings than they were to the microbiomes of other JDM probands. In a sibling-paired within-family analysis, several potentially immunomodulatory bacterial taxa were differentially abundant in the microbiomes of JDM probands compared to their unaffected siblings, including Faecalibacterium (gut) and Streptococcus (oral cavity). While microbiome features of JDM are often shared by unaffected family members, the loss or gain of specific fecal and oral bacteria may play a role in disease pathogenesis or be secondary to immune dysfunction in susceptible individuals.
Collapse
Affiliation(s)
- Sean T Koester
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Kansas School of Medicine, Kansas City, USA
| | - Albert Chow
- Department of Pediatrics, Division of Rheumatology, University of Washington, Seattle, WA, USA
- Loma Linda University, Loma Linda, USA
| | - Evan Pepper-Tunick
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
| | - Peggy Lee
- School of Dentistry, University of Washington, Seattle, WA, USA
| | - Mary Eckert
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA, USA
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Laurie Brenchley
- Office of the Clinical Director, NIDCR, National Institutes of Health, Bethesda, MD, USA
| | - Pamela Gardner
- Office of the Clinical Director, NIDCR, National Institutes of Health, Bethesda, MD, USA
- Oral Oncology at BC Cancer, Vancouver, BC, Canada
| | - Hyun Jung Song
- School of Dentistry, University of Washington, Seattle, WA, USA
| | - Naisi Li
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Adam Schiffenbauer
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Rita Volochayev
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Nastaran Bayat
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
- Social and Scientific Systems, Inc., A DLH Holdings Corp. Company, Silver Spring, MD, USA
| | - Jeffrey S McLean
- Department of Periodontics, University of Washington, Seattle, WA, USA
| | - Lisa G Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Susan Shenoi
- Department of Pediatrics, Division of Rheumatology, University of Washington, Seattle, WA, USA
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Anne M Stevens
- Department of Pediatrics, Division of Rheumatology, University of Washington, Seattle, WA, USA
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA, USA
- Janssen, a Wholly Owned Subsidiary of Johnson & Johnson, Raritan, USA
| | - Neelendu Dey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Medicine, Division of Gastroenterology, University of Washington, Seattle, WA, USA.
- Microbiome Research Initiative, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
36
|
Cao X, Fang Y, Bandan P, Suo L, Jiacuo G, Wu Y, Cuoji A, Zhuoga D, Chen Y, Ji D, Quzhen C, Zhang K. Age-specific composition of milk microbiota in Tibetan sheep and goats. Appl Microbiol Biotechnol 2024; 108:411. [PMID: 38980443 PMCID: PMC11233330 DOI: 10.1007/s00253-024-13252-w] [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: 01/01/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024]
Abstract
This study investigates the dynamic changes in milk nutritional composition and microbial communities in Tibetan sheep and goats during the first 56 days of lactation. Milk samples were systematically collected at five time points (D0, D7, D14, D28, D56) post-delivery. In Tibetan sheep, milk fat, protein, and casein contents were highest on D0, gradually decreased, and stabilized after D14, while lactose and galactose levels showed the opposite trend. Goat milk exhibited similar initial peaks, with significant changes particularly between D0, D7, D14, and D56. 16S rRNA gene sequencing revealed increasing microbial diversity in both species over the lactation period. Principal coordinates analysis identified distinct microbial clusters corresponding to early (D0-D7), transitional (D14-D28), and mature (D56) stages. Core phyla, including Proteobacteria, Firmicutes, Bacteroidetes, and Actinobacteria, dominated the milk microbiota, with significant temporal shifts. Core microbes like Lactobacillus, Leuconostoc, and Streptococcus were common in both species, with species-specific taxa observed (e.g., Pediococcus in sheep, Shewanella in goats). Furthermore, we observed a highly shared core microbiota in sheep and goat milk, including Lactobacillus, Leuconostoc, and Streptococcus. Spearman correlation analysis highlighted significant relationships between specific microbial genera and milk nutrients. For instance, Lactobacillus positively correlated with total solids, non-fat milk solids, protein, and casein, while Mannheimia negatively correlated with protein content. This study underscores the complex interplay between milk composition and microbial dynamics in Tibetan sheep and goats, informing strategies for livestock management and nutritional enhancement. KEY POINTS: • The milk can be classified into three types based on the microbiota composition • The changes of milk microbiota are closely related to the variations in nutrition • Filter out microbiota with species specificity and age specificity in the milk.
Collapse
Affiliation(s)
- Xi Cao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, 712100, Yangling, China
| | - Yumeng Fang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, 712100, Yangling, China
| | - Pingcuo Bandan
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Langda Suo
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Gesang Jiacuo
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Yujiang Wu
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Awang Cuoji
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Deqing Zhuoga
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
| | - Yulin Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, 712100, Yangling, China
| | - De Ji
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China.
| | - Ciren Quzhen
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China.
| | - Ke Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, 712100, Yangling, China.
| |
Collapse
|
37
|
Aktas B, Aslim B, Ozdemir DA. A neurotherapeutic approach with Lacticaseibacillus rhamnosus E9 on gut microbiota and intestinal barrier in MPTP-induced mouse model of Parkinson's disease. Sci Rep 2024; 14:15460. [PMID: 38965287 PMCID: PMC11224381 DOI: 10.1038/s41598-024-65061-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024] Open
Abstract
The gut microbiota plays a crucial role in neural development and progression of neural disorders like Parkinson's disease (PD). Probiotics have been suggested to impact neurodegenerative diseases via gut-brain axis. This study aims to investigate the therapeutic potential of Lacticaseibacillus rhamnosus E9, a high exopolysaccharide producer, on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine(MPTP)-induced mouse model of PD. C57BL/6 mice subjected to MPTP were fed L. rhamnosus E9 for fifteen days and sacrificed after the last administration. Motor functions were determined by open-field, catalepsy, and wire-hanging tests. The ileum and the brain tissues were collected for ELISA, qPCR, and immunohistochemistry analyses. The cecum content was obtained for microbiota analysis. E9 supplementation alleviated MPTP-induced motor dysfunctions accompanied by decreased levels of striatal TH and dopamine. E9 also reduced the level of ROS in the striatum and decreased the DAT expression while increasing the DR1. Furthermore, E9 improved intestinal integrity by enhancing ZO-1 and Occludin levels and reversed the dysbiosis of the gut microbiota induced by MPTP. In conclusion, E9 supplementation improved the MPTP-induced motor deficits and neural damage as well as intestinal barrier by modulating the gut microbiota in PD mice. These findings suggest that E9 supplementation holds therapeutic potential in managing PD through the gut-brain axis.
Collapse
Affiliation(s)
- Busra Aktas
- Department of Molecular Biology and Genetics, Burdur Mehmet Akif Ersoy University, Burdur, 15200, Turkey.
| | - Belma Aslim
- Department of Biology, Faculty of Science, Gazi University, Ankara, 06500, Turkey
| | - Deniz Ates Ozdemir
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, 06230, Turkey
| |
Collapse
|
38
|
Hasnain MA, Kang D, Moon GS. Research trends of next generation probiotics. Food Sci Biotechnol 2024; 33:2111-2121. [PMID: 39130671 PMCID: PMC11315851 DOI: 10.1007/s10068-024-01626-9] [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: 04/08/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 08/13/2024] Open
Abstract
Gut represents one of the largest interfaces for the interaction of host factors and the environmental ones. Gut microbiota, largely dominated by bacterial community, plays a significant role in the health status of the host. The healthy gut microbiota fulfills several vital functions such as energy metabolism, disease protection, and immune modulation. Dysbiosis, characterized by microbial imbalance, can contribute to the development of various disorders, including intestinal, systemic, metabolic, and neurodegenerative conditions. Probiotics offer the potential to address dysbiosis and improve overall health. Advancements in high-throughput sequencing, bioinformatics, and omics have enabled mechanistic studies for the development of bespoke probiotics, referred to as next generation probiotics. These tailor-made probiotics have the potential to ameliorate specific disease conditions and thus fulfill the specific consumer needs. This review discusses recent updates on the most promising next generation probiotics, along with the challenges that must be addressed to translate this concept into reality.
Collapse
Affiliation(s)
- Muhammad Adeel Hasnain
- Major in IT·Biohealth Convergence, Department of IT·Energy Convergence, Graduate School, Korea National University of Transportation, Chungju, 27469 Republic of Korea
| | - Dae‑Kyung Kang
- Department of Animal Resources Science, Dankook University, Cheonan, 31116 Republic of Korea
| | - Gi-Seong Moon
- Major in IT·Biohealth Convergence, Department of IT·Energy Convergence, Graduate School, Korea National University of Transportation, Chungju, 27469 Republic of Korea
- Major in Biotechnology, Korea National University of Transportation, Jeungpyeong, 27909 Republic of Korea
- 4D Convergence Technology Institute, Korea National University of Transportation, Jeungpyeong, 27909 Republic of Korea
| |
Collapse
|
39
|
Chen K, Wang YC, Yang KT, Huang TH, Hung YM, Chang R. Association of diverticulitis and potential risk of ischemic stroke: population-based matched cohort study in Taiwan. Postgrad Med J 2024; 100:475-481. [PMID: 38453141 DOI: 10.1093/postmj/qgae010] [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: 09/14/2023] [Revised: 11/20/2023] [Accepted: 12/02/2023] [Indexed: 03/09/2024]
Abstract
BACKGROUND Previous studies have suggested relationship between diverticular disease and cardiovascular disease. Since cardiovascular disease and cerebrovascular accident share a lot of pathogenesis, diverticulitis could also be a risk factor for stroke. This study tried to establish epidemiological evidence of the relationship between colon diverticulitis and ischemic stroke. METHODS In this retrospective cohort study, patients with newly diagnosed colon diverticulitis (N = 6238) and patients without colon diverticulitis (control group; N = 24 952) were recruited between January 1, 2000, and December 31, 2017. Both groups were matched by propensity score at a 1:4 ratio by age, sex, comorbidities and medications. Cox proportional hazard regression was applied to estimate the hazard ratio (HR) and 95% confidence interval (CI) of ischemic stroke. We also conducted 4 different regression models and 2 sensitivity analyses to test the robustness of our findings. RESULTS The diverticulitis group had a higher risk of IS than the control group (adjusted HR, 1.25; 95% CI, 1.12-1.39; P < 0.001). Serial sensitivity analyses yielded consistent positive link between diverticulitis and IS. Further subgroup analysis showed that in the study group, the risk of IS was 2.54-fold higher than the matched controls in 30-39 years. CONCLUSIONS Our study found that colon diverticulitis was associated with a higher risk of developing subsequent ischemic stroke, especially for patients aged 30-39 years, among Asian population. This result provides us a chance to undertake preventive measures for ischemic stroke in high-risk patients.
Collapse
Affiliation(s)
- Kuan Chen
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| | - Yung-Chang Wang
- Department of Surgery, Kaohsiung Municipal Min-Sheng Hospital, Kaohsiung 813, Taiwan
- Graduate Institute of Adult Education, College of Education, National Kaohsiung Normal University, Kaohsiung 813, Taiwan
| | - Kuang-Tsu Yang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Municipal Min-Sheng Hospital, Kaohsiung 813, Taiwan
| | - Ting-Hsin Huang
- Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| | - Yao-Min Hung
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital Taitung Branch, Taitung 950, Taiwan
- College of Science and Engineering, National Taitung University, Taitung 950, Taiwan
- College of Health and Nursing, Meiho University, Pingtung 912, Taiwan
| | - Renin Chang
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| |
Collapse
|
40
|
Zhang K, Nakaoka S. An energy landscape approach reveals the potential key bacteria contributing to the development of inflammatory bowel disease. PLoS One 2024; 19:e0302151. [PMID: 38885178 PMCID: PMC11182530 DOI: 10.1371/journal.pone.0302151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 03/28/2024] [Indexed: 06/20/2024] Open
Abstract
The dysbiosis of microbiota has been reported to be associated with numerous human pathophysiological processes, including inflammatory bowel disease (IBD). With advancements in high-throughput sequencing, various methods have been developed to study the alteration of microbiota in the development and progression of diseases. However, a suitable approach to assess the global stability of the microbiota in disease states through time-series microbiome data is yet to be established. In this study, we have introduced a novel Energy Landscape construction method, which incorporates the Latent Dirichlet Allocation (LDA) model and the pairwise Maximum Entropy (MaxEnt) model for their complementary advantages, and demonstrate its utility by applying it to an IBD time-series dataset. Through this approach, we obtained the microbial assemblages' energy profile of the whole microbiota under the IBD condition and uncovered the hidden stable stages of microbiota structure during the disease development with time-series microbiome data. The Bacteroides-dominated assemblages presenting in multiple stable states suggest the potential contribution of Bacteroides and interactions with other microbial genera, like Alistipes, and Faecalibacterium, to the development of IBD. Our proposed method provides a novel and insightful tool for understanding the alteration and stability of the microbiota under disease states and offers a more holistic view of the complex dynamics at play in microbiota-mediated diseases.
Collapse
Affiliation(s)
- Kaiyang Zhang
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Shinji Nakaoka
- Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
41
|
Liang Y, Chang Y, Xie Y, Hou Q, Zhao H, Liu G, Chen X, Tian G, Cai J, Jia G. Dietary ethylenediamine dihydroiodide mitigated Escherichia coli O78-induced immune and intestinal damage of ducks via suppression of NF-κB signal. Poult Sci 2024; 103:103610. [PMID: 38489887 PMCID: PMC10952079 DOI: 10.1016/j.psj.2024.103610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/17/2024] [Accepted: 02/29/2024] [Indexed: 03/17/2024] Open
Abstract
This study investigated the effect of Ethylenediamine dihydroiodide (EDDI) on growth performance, immune function and intestinal health of meat ducks challenged with Avian pathogenic Escherichia coli (APEC). A total of 360 one-day-old Cherry Valley ducks with similar body weight were randomly allocated to 6 treatments (6 floor cages, 10 birds/cage). A 3 × 2 factor design was used with 3 dietary iodine levels (0, 8, 16 mg/kg in the form EDDI and whether APEC was challenged or not at 7-day-old ducks. The feeding period lasted for 20 d. The results showed that the addition of EDDI reduced APEC-induced decrease of the 20-d weight loss of meat ducks (P < 0.05), and alleviated the inflammatory response of liver tissue induced by APEC challenge in meat ducks. In terms of immune function, EDDI supplementation reduced the immune organ index and increased the immune cell count of meat ducks, reduced the level of endotoxins in the serum of meat ducks (P < 0.05), as well as inhibited the expression levels of liver and spleen inflammatory factors and TLR signaling pathway related genes induced by APEC (P < 0.05). In terms of intestinal health, EDDI inhibited APEC-induced decreases in ZO-3 genes expression and increases in IL-1β and TNF-α expression, increased relative abundance of beneficial bacteria in the cecum and content of metabolites. Pearson correlation analysis showed that there was a significant correlation between liver inflammatory factors and TLR4 signaling pathway genes, and there might be a significant correlation between intestinal microbial flora and other physiological indexes of meat ducks, which indicated that EDDI could reduce the damage to immune function and intestinal health caused by APEC challenge through regulating the structure of intestinal flora. Collectively, our findings suggest that the EDDI can promote growth performance, improve immune function and the intestinal barrier in APEC-challenged meat ducks, which may be related to the suppression of NF-κB signal.
Collapse
Affiliation(s)
- Yanru Liang
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Yaqi Chang
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Yueqin Xie
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Qinteng Hou
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Hua Zhao
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Guangmang Liu
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Xiaoling Chen
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Gang Tian
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jingyi Cai
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Gang Jia
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
42
|
Crouch LI, Rodrigues CS, Bakshani CR, Tavares-Gomes L, Gaifem J, Pinho SS. The role of glycans in health and disease: Regulators of the interaction between gut microbiota and host immune system. Semin Immunol 2024; 73:101891. [PMID: 39388764 DOI: 10.1016/j.smim.2024.101891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
The human gut microbiota is home to a diverse collection of microorganisms that has co-evolved with the host immune system in which host-microbiota interactions are essential to preserve health and homeostasis. Evidence suggests that the perturbation of this symbiotic host-microbiome relationship contributes to the onset of major diseases such as chronic inflammatory diseases including Inflammatory Bowel Disease. The host glycocalyx (repertoire of glycans/sugar-chains at the surface of gut mucosa) constitutes a major biological and physical interface between the intestinal mucosa and microorganisms, as well as with the host immune system. Glycans are an essential niche for microbiota colonization and thus an important modulator of host-microorganism interactions both in homeostasis and in disease. In this review, we discuss the role of gut mucosa glycome as an instrumental pathway that regulates host-microbiome interactions in homeostasis but also in health to inflammation transition. We also discuss the power of mucosa glycosylation remodelling as an attractive preventive and therapeutic strategy to preserve gut homeostasis.
Collapse
Affiliation(s)
- Lucy I Crouch
- Department of Microbes, Infection and Microbiomes, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK.
| | - Cláudia S Rodrigues
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
| | - Cassie R Bakshani
- Department of Microbes, Infection and Microbiomes, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Leticia Tavares-Gomes
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Joana Gaifem
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Salomé S Pinho
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
43
|
Kennedy JM, De Silva A, Walton GE, Gibson GR. A review on the use of prebiotics in ulcerative colitis. Trends Microbiol 2024; 32:507-515. [PMID: 38065786 DOI: 10.1016/j.tim.2023.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 05/12/2024]
Abstract
The gut microbiome in the inflammatory bowel disease, ulcerative colitis (UC), is different to that of healthy controls. Patients with UC have relative reductions in abundance of Firmicutes and Bifidobacterium in the colon, and an increase in sulfate-reducing bacteria. Prebiotics are dietary substrates which are selectively metabolised by the human colonic microbiota to confer health benefits to the host. This review explores our current understanding of the potential benefits of prebiotics on various clinical, biochemical, and microbiological endpoints in UC, including new perspectives gained from recent studies in the field. This review looks to the future and highlights the need for appropriately designed trials to explore this potentially exciting new avenue for the treatment of UC.
Collapse
Affiliation(s)
- James M Kennedy
- Department of Food and Nutritional Sciences, The University of Reading, Reading, RG6 6AP, UK; Department of Gastroenterology, Royal Berkshire NHS Foundation Trust, Reading, RG1 5AN, UK.
| | - Aminda De Silva
- Department of Gastroenterology, Royal Berkshire NHS Foundation Trust, Reading, RG1 5AN, UK
| | - Gemma E Walton
- Department of Food and Nutritional Sciences, The University of Reading, Reading, RG6 6AP, UK
| | - Glenn R Gibson
- Department of Food and Nutritional Sciences, The University of Reading, Reading, RG6 6AP, UK
| |
Collapse
|
44
|
Cicchinelli S, Gemma S, Pignataro G, Piccioni A, Ojetti V, Gasbarrini A, Franceschi F, Candelli M. Intestinal Fibrogenesis in Inflammatory Bowel Diseases: Exploring the Potential Role of Gut Microbiota Metabolites as Modulators. Pharmaceuticals (Basel) 2024; 17:490. [PMID: 38675450 PMCID: PMC11053610 DOI: 10.3390/ph17040490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Fibrosis, sustained by the transformation of intestinal epithelial cells into fibroblasts (epithelial-to-mesenchymal transition, EMT), has been extensively studied in recent decades, with the molecular basis well-documented in various diseases, including inflammatory bowel diseases (IBDs). However, the factors influencing these pathways remain unclear. In recent years, the role of the gut microbiota in health and disease has garnered significant attention. Evidence suggests that an imbalanced or dysregulated microbiota, along with environmental and genetic factors, may contribute to the development of IBDs. Notably, microbes produce various metabolites that interact with host receptors and associated signaling pathways, influencing physiological and pathological changes. This review aims to present recent evidence highlighting the emerging role of the most studied metabolites as potential modulators of molecular pathways implicated in intestinal fibrosis and EMT in IBDs. These studies provide a deeper understanding of intestinal inflammation and fibrosis, elucidating the molecular basis of the microbiota role in IBDs, paving the way for future treatments.
Collapse
Affiliation(s)
- Sara Cicchinelli
- Department of Emergency, S.S. Filippo e Nicola Hospital, 67051 Avezzano, Italy;
| | - Stefania Gemma
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giulia Pignataro
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Andrea Piccioni
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Veronica Ojetti
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| |
Collapse
|
45
|
Acar C, Celik SK, Ozdemirel HO, Tuncdemir BE, Alan S, Mergen H. Composition of the colon microbiota in the individuals with inflammatory bowel disease and colon cancer. Folia Microbiol (Praha) 2024; 69:333-345. [PMID: 37344611 DOI: 10.1007/s12223-023-01072-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
The human intestine is a habitat for microorganisms and, recently, the composition of the intestinal microbiota has been correlated with the etiology of diseases such as inflammations, sores, and tumors. Although many studies have been conducted to understand the composition of that microbiota, expanding these studies to more samples and different backgrounds will improve our knowledge. In this work, we showed the colon microbiota composition and diversity of healthy subjects, patients with inflammatory bowel disease (IBD), and colon cancer by metagenomic sequencing. Our results indicated that the relative abundance of prokaryotic and eukaryotic microbes differs between the healthy vs. tumor biopsies, tumor vs. IBD biopsies, and fresh vs. paraffin-embedded tumor biopsies. Fusobacterium, Escherichia-Shigella, and Streptococcus genera were relatively abundant in fresh tumor biopsies, while Pseudomonas was significantly elevated in IBD biopsies. Additionally, another opportunist pathogen Malasseziales was revealed as the most abundant fungal clade in IBD biopsies, especially in ulcerative colitis. We also found that, while the Basidiomycota:Ascomycota ratio was slightly lower in tumor biopsies compared to biopsies from healthy subjects, there was a significant increase in IBD biopsies. Our work will contribute to the known diversity of prokaryotic and eukaryotic microbes in the colon biopsies in patients with IBD and colon cancer.
Collapse
Affiliation(s)
- Ceren Acar
- Faculty of Science and Literature, Department of Molecular Biology and Genetics, Inonu University, Malatya, 44280, Turkey.
| | | | - H Ozgur Ozdemirel
- Faculty of Science, Department of Biology, Hacettepe University, Ankara, 06800, Turkey
| | - Beril Erdem Tuncdemir
- Faculty of Science, Department of Biology, Hacettepe University, Ankara, 06800, Turkey
| | - Saadet Alan
- Faculty of Medicine, Department of Medical Pathology, Inonu University, Malatya, 44000, Turkey
| | - Hatice Mergen
- Faculty of Science, Department of Biology, Hacettepe University, Ankara, 06800, Turkey
| |
Collapse
|
46
|
Williams LM, Cao S. Harnessing and delivering microbial metabolites as therapeutics via advanced pharmaceutical approaches. Pharmacol Ther 2024; 256:108605. [PMID: 38367866 PMCID: PMC10985132 DOI: 10.1016/j.pharmthera.2024.108605] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/08/2024] [Indexed: 02/19/2024]
Abstract
Microbial metabolites have emerged as key players in the interplay between diet, the gut microbiome, and host health. Two major classes, short-chain fatty acids (SCFAs) and tryptophan (Trp) metabolites, are recognized to regulate inflammatory, immune, and metabolic responses within the host. Given that many human diseases are associated with dysbiosis of the gut microbiome and consequent reductions in microbial metabolite production, the administration of these metabolites represents a direct, multi-targeted treatment. While a multitude of preclinical studies showcase the therapeutic potential of both SCFAs and Trp metabolites, they often rely on high doses and frequent dosing regimens to achieve systemic effects, thereby constraining their clinical applicability. To address these limitations, a variety of pharmaceutical formulations approaches that enable targeted, delayed, and/or sustained microbial metabolite delivery have been developed. These approaches, including enteric encapsulations, esterification to dietary fiber, prodrugs, and nanoformulations, pave the way for the next generation of microbial metabolite-based therapeutics. In this review, we first provide an overview of the roles of microbial metabolites in maintaining host homeostasis and outline how compromised metabolite production contributes to the pathogenesis of inflammatory, metabolic, autoimmune, allergic, infectious, and cancerous diseases. Additionally, we explore the therapeutic potential of metabolites in these disease contexts. Then, we provide a comprehensive and up-to-date review of the pharmaceutical strategies that have been employed to enhance the therapeutic efficacy of microbial metabolites, with a focus on SCFAs and Trp metabolites.
Collapse
Affiliation(s)
- Lindsey M Williams
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States
| | - Shijie Cao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
47
|
Hong J, Fu T, Liu W, Du Y, Bu J, Wei G, Yu M, Lin Y, Min C, Lin D. An Update on the Role and Potential Molecules in Relation to Ruminococcus gnavus in Inflammatory Bowel Disease, Obesity and Diabetes Mellitus. Diabetes Metab Syndr Obes 2024; 17:1235-1248. [PMID: 38496006 PMCID: PMC10942254 DOI: 10.2147/dmso.s456173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/27/2024] [Indexed: 03/19/2024] Open
Abstract
Ruminococcus gnavus (R. gnavus) is a gram-positive anaerobe commonly resides in the human gut microbiota. The advent of metagenomics has linked R. gnavus with various diseases, including inflammatory bowel disease (IBD), obesity, and diabetes mellitus (DM), which has become a growing area of investigation. The initial focus of research primarily centered on assessing the abundance of R. gnavus and its potential association with disease presentation, taking into account variations in sample size, sequencing and analysis methods. However, recent investigations have shifted towards elucidating the underlying mechanistic pathways through which R. gnavus may contribute to disease manifestation. In this comprehensive review, we aim to provide an updated synthesis of the current literature on R. gnavus in the context of IBD, obesity, and DM. We critically analyze relevant studies and summarize the potential molecular mediators implicated in the association between R. gnavus and these diseases. Across numerous studies, various molecules such as methylation-controlled J (MCJ), glucopolysaccharides, ursodeoxycholic acid (UDCA), interleukin(IL)-10, IL-17, and capric acid have been proposed as potential contributors to the link between R. gnavus and IBD. Similarly, in the realm of obesity, molecules such as hydrogen peroxide, butyrate, and UDCA have been suggested as potential mediators, while glycine ursodeoxycholic acid (GUDCA) has been implicated in the connection between R. gnavus and DM. Furthermore, it is imperative to emphasize the necessity for additional studies to evaluate the potential efficacy of targeting pathways associated with R. gnavus as a viable strategy for managing these diseases. These findings have significantly expanded our understanding of the functional role of R. gnavus in the context of IBD, obesity, and DM. This review aims to offer updated insights into the role and potential mechanisms of R. gnavus, as well as potential strategies for the treatment of these diseases.
Collapse
Affiliation(s)
- Jinni Hong
- Department of Traditional Chinese Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Guangdong Provincial Institute of Geriatric, Guangzhou, Guangdong, 510080, People’s Republic of China
| | - Tingting Fu
- Department of Traditional Chinese Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Guangdong Provincial Institute of Geriatric, Guangzhou, Guangdong, 510080, People’s Republic of China
| | - Weizhen Liu
- Department of Traditional Chinese Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Guangdong Provincial Institute of Geriatric, Guangzhou, Guangdong, 510080, People’s Republic of China
| | - Yu Du
- Department of Traditional Chinese Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Guangdong Provincial Institute of Geriatric, Guangzhou, Guangdong, 510080, People’s Republic of China
| | - Junmin Bu
- Department of Traditional Chinese Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Guangdong Provincial Institute of Geriatric, Guangzhou, Guangdong, 510080, People’s Republic of China
| | - Guojian Wei
- Department of Traditional Chinese Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Guangdong Provincial Institute of Geriatric, Guangzhou, Guangdong, 510080, People’s Republic of China
| | - Miao Yu
- Department of Traditional Chinese Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Guangdong Provincial Institute of Geriatric, Guangzhou, Guangdong, 510080, People’s Republic of China
| | - Yanshan Lin
- Department of Traditional Chinese Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Guangdong Provincial Institute of Geriatric, Guangzhou, Guangdong, 510080, People’s Republic of China
| | - Cunyun Min
- Department of Traditional Chinese Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Guangdong Provincial Institute of Geriatric, Guangzhou, Guangdong, 510080, People’s Republic of China
| | - Datao Lin
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, People’s Republic of China
| |
Collapse
|
48
|
Chen S, Mei H, Xu L, Zhan L, Yang Y, Zhao D, Bao G, Li X, Cao Z. Impact of fermented feed of soybean hulls and rapeseed cake on immunity, antioxidant capacity, and gut microbiota in Chahua chicken. Poult Sci 2024; 103:103451. [PMID: 38301497 PMCID: PMC10847688 DOI: 10.1016/j.psj.2024.103451] [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: 10/31/2023] [Revised: 12/25/2023] [Accepted: 01/06/2024] [Indexed: 02/03/2024] Open
Abstract
The present study investigated the effects of replacing part of the basal diet with 2-stage fermented feed (FF) (soybean hulls:rapeseed cake (2:1, m/m)) on the growth performance, immunity, antioxidant capacity, and intestinal health of Chahua chicken. A total of 160 Chahua chickens were randomly divided into 4 groups to receive a control diet or diet with 5%, 10%, or 15% of the basal diet replaced by FF, respectively for 56 d. The results showed that FF significantly improved the average daily gain (ADG) and average daily feed intake (ADFI) of Chahua chickens (P < 0.05). Furthermore, the serum immunoglobulin (Ig) A, glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD) in Chahua chicken receiving the diet added with 15% FF significantly increased (P < 0.05). Chahua chicken in both the 10% and 15% groups showed increased serum IgG and IgM and decreased malondialdehyde. Serum interleukin-2 and interferon-gamma significantly increased in all FF groups. Compared with the CON group, higher ileal villus height (VH) was found in the 10% FF group. Treatment with FF significantly increased the ileal villus height/crypt depth (VH/CD) ratio, jejunal VH, and jejunal VH/CD ratio while reducing ileal and jejunal CD. The modified gut microbiota composition was observed in the Chahua chicken fed a diet containing FF, in particular, with the increased abundance of Faecalibacterium and Lactobacillus. The abundance of Lactobacillus significantly increased in the 10% and 15% FF groups (all P < 0.05). Correlation analysis revealed a positive correlation between Lactobacillus and VH (R = 0.38, P = 0.10, Figure 3B), AH/CD ratio (R = 0.63, P = 0.003), and a negative correlation with CD (R = -0.72, P = 0.001). These results indicate that FF improves immunity, antioxidant capacity, and intestinal health and consequently enhances growth performance in Chahua chicken.
Collapse
Affiliation(s)
- Shiyu Chen
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China
| | - Huiyou Mei
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China
| | - Le Xu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China; Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Heilongtan, Kunming 650201, People's Republic of China
| | - Limei Zhan
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China
| | - Yuhao Yang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China
| | - Dexuan Zhao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China
| | - Guoying Bao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China
| | - Xiaoye Li
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China
| | - Zhenhui Cao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China; Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Heilongtan, Kunming 650201, People's Republic of China.
| |
Collapse
|
49
|
Danne C, Skerniskyte J, Marteyn B, Sokol H. Neutrophils: from IBD to the gut microbiota. Nat Rev Gastroenterol Hepatol 2024; 21:184-197. [PMID: 38110547 DOI: 10.1038/s41575-023-00871-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2023] [Indexed: 12/20/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal tract that results from dysfunction in innate and/or adaptive immune responses. Impaired innate immunity, which leads to lack of control of an altered intestinal microbiota and to activation of the adaptive immune system, promotes a secondary inflammatory response that is responsible for tissue damage. Neutrophils are key players in innate immunity in IBD, but their roles have been neglected compared with those of other immune cells. The latest studies on neutrophils in IBD have revealed unexpected complexities, with heterogeneous populations and dual functions, both deleterious and protective, for the host. In parallel, interconnections between disease development, intestinal microbiota and neutrophils have been highlighted. Numerous IBD susceptibility genes (such as NOD2, NCF4, LRRK2, CARD9) are involved in neutrophil functions related to defence against microorganisms. Moreover, severe monogenic diseases involving dysfunctional neutrophils, including chronic granulomatous disease, are characterized by intestinal inflammation that mimics IBD and by alterations in the intestinal microbiota. This observation demonstrates the dialogue between neutrophils, gut inflammation and the microbiota. Neutrophils affect microbiota composition and function in several ways. In return, microbial factors, including metabolites, regulate neutrophil production and function directly and indirectly. It is crucial to further investigate the diverse roles played by neutrophils in host-microbiota interactions, both at steady state and in inflammatory conditions, to develop new IBD therapies. In this Review, we discuss the roles of neutrophils in IBD, in light of emerging evidence proving strong interconnections between neutrophils and the gut microbiota, especially in an inflammatory context.
Collapse
Affiliation(s)
- Camille Danne
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Gastroentérologie, Paris, France.
- Paris Center For Microbiome Medicine (PaCeMM) FHU, Paris, France.
| | - Jurate Skerniskyte
- CNRS, UPR 9002, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire, Architecture et Réactivité de l'ARN, Strasbourg, France
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Benoit Marteyn
- CNRS, UPR 9002, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire, Architecture et Réactivité de l'ARN, Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France
- Institut Pasteur, Université de Paris, Inserm 1225 Unité de Pathogenèse des Infections Vasculaires, Paris, France
| | - Harry Sokol
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Gastroentérologie, Paris, France
- Paris Center For Microbiome Medicine (PaCeMM) FHU, Paris, France
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| |
Collapse
|
50
|
Alves Costa Silva C, Piccinno G, Suissa D, Bourgin M, Schreibelt G, Durand S, Birebent R, Fidelle M, Sow C, Aprahamian F, Manghi P, Punčochář M, Asnicar F, Pinto F, Armanini F, Terrisse S, Routy B, Drubay D, Eggermont AMM, Kroemer G, Segata N, Zitvogel L, Derosa L, Bol KF, de Vries IJM. Influence of microbiota-associated metabolic reprogramming on clinical outcome in patients with melanoma from the randomized adjuvant dendritic cell-based MIND-DC trial. Nat Commun 2024; 15:1633. [PMID: 38395948 PMCID: PMC10891084 DOI: 10.1038/s41467-024-45357-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor immunosurveillance plays a major role in melanoma, prompting the development of immunotherapy strategies. The gut microbiota composition, influencing peripheral and tumoral immune tonus, earned its credentials among predictors of survival in melanoma. The MIND-DC phase III trial (NCT02993315) randomized (2:1 ratio) 148 patients with stage IIIB/C melanoma to adjuvant treatment with autologous natural dendritic cell (nDC) or placebo (PL). Overall, 144 patients collected serum and stool samples before and after 2 bimonthly injections to perform metabolomics (MB) and metagenomics (MG) as prespecified exploratory analysis. Clinical outcomes are reported separately. Here we show that different microbes were associated with prognosis, with the health-related Faecalibacterium prausnitzii standing out as the main beneficial taxon for no recurrence at 2 years (p = 0.008 at baseline, nDC arm). Therapy coincided with major MB perturbations (acylcarnitines, carboxylic and fatty acids). Despite randomization, nDC arm exhibited MG and MB bias at baseline: relative under-representation of F. prausnitzii, and perturbations of primary biliary acids (BA). F. prausnitzii anticorrelated with BA, medium- and long-chain acylcarnitines. Combined, these MG and MB biomarkers markedly determined prognosis. Altogether, the host-microbial interaction may play a role in localized melanoma. We value systematic MG and MB profiling in randomized trials to avoid baseline differences attributed to host-microbe interactions.
Collapse
Grants
- The MIND-DC trial was funded by ZonMw, Ministry of Health, Welfare and Sport (VWS), Stichting ATK, Miltenyi Biotec (in-kind). This work was supported by SEERAVE Foundation, European Union Horizon 2020:Project Number: 825410 and Project Acronym: ONCOBIOME, Institut National du Cancer (INCa), ANR Ileobiome - 19-CE15-0029-01, ANR RHU5 “ANR-21-RHUS-0017” IMMUNOLIFE&#x201D;, MAdCAM INCA_ 16698, Ligue contre le cancer, LABEX OncoImmunology, la direction generale de l&#x2019;offre de soins (DGOS), Universite Paris-Sud, SIRIC SOCRATE (INCa/DGOS/INSERM 6043), and PACRI network. G.K. is supported by the Ligue contre le Cancer (équipe labellis&#x00E9;e); Agence National de la Recherche (ANR) – Projets blancs; AMMICa US23/CNRS UMS3655; Association pour la recherche sur le cancer (ARC); Canc&#x00E9;rop&#x00F4;le Ile-de-France; Fondation pour la Recherche M&#x00E9;dicale (FRM); a donation by Elior; Equipex Onco-Pheno-Screen; European Joint Programme on Rare Diseases (EJPRD); European Research Council Advanced Investigator Award (ERC-2021-ADG, ICD-Cancer, Grant No. 101052444), European Union Horizon 2020 Projects Oncobiome, Prevalung (grant No. 101095604) and Crimson; Fondation Carrefour; Institut National du Cancer (INCa); Institut Universitaire de France; LabEx Immuno-Oncology (ANR-18-IDEX-0001); a Cancer Research ASPIRE Award from the Mark Foundation; the RHU Immunolife; Seerave Foundation; SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); and SIRIC Cancer Research and Personalized Medicine (CARPEM). This study contributes to the IdEx Universit&#x00E9; de Paris ANR-18-IDEX-0001. This work is supported by the Prism project funded by the Agence Nationale de la Recherche under grant number ANR-18-IBHU-0002. CACS was funded by MSD Avenir. MF is funded by SEERAVE Foundation and MERCK Foundation. LD and BR were supported by Philantropia at Gustave Roussy Foundation.
Collapse
Affiliation(s)
- Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Gianmarco Piccinno
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Déborah Suissa
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Mélanie Bourgin
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
| | - Gerty Schreibelt
- Medical BioSciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Sylvère Durand
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
| | - Roxanne Birebent
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Marine Fidelle
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Cissé Sow
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Fanny Aprahamian
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
| | - Paolo Manghi
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Michal Punčochář
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Francesco Asnicar
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Federica Pinto
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Federica Armanini
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Safae Terrisse
- Oncology Department, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis, Paris, France
| | - Bertrand Routy
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Hematology-Oncology Division, Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Damien Drubay
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Office of Biostatistics and Epidemiology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Inserm, Université Paris-Saclay, CESP U1018, Oncostat, labeled Ligue Contre le Cancer, Villejuif, France
| | - Alexander M M Eggermont
- Princess Máxima Center and University Medical Center Utrecht, 3584 CS Utrecht, The Netherlands
- Comprehensive Cancer Center Munich, Technical University Munich & Ludwig Maximiliaan University, Munich, Germany
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Nicola Segata
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France.
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France.
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France.
- Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, Villejuif, France.
| | - Lisa Derosa
- Gustave Roussy Cancer Campus (GRCC), ClinicObiome, Villejuif Cedex, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Kalijn F Bol
- Medical BioSciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Medical BioSciences, Radboud Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|