1
|
Nichols L, El-Kholy O, Elsayed AAR, Basson MD. The bidirectional interplay between gut dysbiosis and surgical complications: A systematic review. Am J Surg 2025; 245:116369. [PMID: 40344995 DOI: 10.1016/j.amjsurg.2025.116369] [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: 03/30/2025] [Revised: 04/16/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND The gut microbiome critically influences diverse aspects of physiology and surgical recovery. Conversely, surgery alters the microbiome, potentially predisposing to complications. We aimed to clarify the bidirectional interaction between surgery and gut dysbiosis. METHODS On December 22nd, 2024, a systematic search of the Cochrane Library, PubMed, VHL, and WOS was completed. Relevant studies were assessed for risk of bias using STROBE and CONSORT guidelines. RESULTS Thirty studies, with 2500+ participants experiencing diverse procedures and complications, were incorporated. Although specifics varied, dysbiosis correlated with surgery and its complications. Patients with complications had more harmful bacteria and fewer beneficial bacteria. In some studies, probiotics reduced complications. CONCLUSION Gut dysbiosis is tied to postoperative complications in a complex, bidirectional relationship. Patients with surgical complications may have fewer beneficial and more pathogenic bacteria both before and after surgery. Early identification of dysbiosis and probiotic administration could predict or even reduce postoperative complications.
Collapse
Affiliation(s)
- Lindsey Nichols
- College of Medicine, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Omar El-Kholy
- Faculty of Medicine, Alexandria University, Alexandria, 21521, Egypt
| | - Ahmed Adham R Elsayed
- College of Medicine, Northeast Ohio Medical University, Rootstown, OH, 44272, USA; Department of Surgery, Northeast Ohio Medical University, Rootstown, OH, 44272, USA; Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Marc D Basson
- College of Medicine, Northeast Ohio Medical University, Rootstown, OH, 44272, USA; Department of Surgery, Northeast Ohio Medical University, Rootstown, OH, 44272, USA; Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, 44272, USA.
| |
Collapse
|
2
|
Zhang Z, Wang J, Dang S, Liu X, Zhang Y, Zhang H. The worldview of Akkermansia muciniphila, a bibliometric analysis. Front Microbiol 2025; 16:1500893. [PMID: 40104597 PMCID: PMC11913835 DOI: 10.3389/fmicb.2025.1500893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/13/2025] [Indexed: 03/20/2025] Open
Abstract
Akkermansia muciniphila (A. muciniphila), a critical bacterium within the gut microbiota, plays a key role in human health and immunomodulation. Since its identification in 2004, A. muciniphila has emerged as a significant agent in treating metabolic diseases, gastroenterological diseases, and tumor immunotherapy. Its rapid ascent in scientific translation underscores its importance in gut microbiome research. However, there has been a lack of visualization and analysis of the rapidly occurring commercialization in this field, which has critically hindered insights into the current knowledge structure and understanding of the cutting-edge of the discipline. This study employs the Web of Science Core Collection (WOSCC) and Innography platforms to provide the first comprehensive analysis of A. muciniphila's academic progresses and commercialization over the past two decades, highlighting its growing prominence in global health research. Our analysis delineates that, following the academic trajectory, the evolution of A. muciniphila patents from foundational research through to application development and maturity, with particular emphasis on its expansive potential in emerging fields, including gastroenterological disorders, non-alcoholic fatty liver disease, cancer immunotherapy, stress management, and neurodegenerative disease treatment. Concluding, A. muciniphila presents as a next-generation probiotic with vast implications for human health. Our findings provide essential insights for future research and product development, contributing to the advancement of this burgeoning field.
Collapse
Affiliation(s)
- Zhao Zhang
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyu Wang
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Shaoqing Dang
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xingzi Liu
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuemiao Zhang
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Ryan N, O’Mahony S, Leahy-Warren P, Philpott L, Mulcahy H. The impact of perinatal maternal stress on the maternal and infant gut and human milk microbiomes: A scoping review. PLoS One 2025; 20:e0318237. [PMID: 40019912 PMCID: PMC11870360 DOI: 10.1371/journal.pone.0318237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/14/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Perinatal maternal stress, which includes both psychological and physiological stress experienced by healthy women during pregnancy and the postpartum period, is becoming increasingly prevalent. Infant early exposure to adverse environments such as perinatal stress has been shown to increase the long-term risk to metabolic, immunologic and neurobehavioral disorders. Evidence suggests that the human microbiome facilitates the transmission of maternal factors to infants via the vaginal, gut, and human milk microbiomes. The colonization of aberrant microorganisms in the mother's microbiome, influenced by the microbiome-brain-gut axis, may be transferred to infants during a critical early developmental period. This transfer may predispose infants to a more inflammatory-prone microbiome which is associated with dysregulated metabolic process leading to adverse health outcomes. Given the prevalence and potential impact of perinatal stress on maternal and infant health, with no systematic mapping or review of the data to date, the aim of this scoping review is to gather evidence on the relationship between perinatal maternal stress, and the human milk, maternal, and infant gut microbiomes. METHODS This is an exploratory mapping scoping review, guided by the Joanna Briggs Institute's methodology along with use of the Prisma Scr reporting guideline. A comprehensive search was conducted using the following databases, CINAHL Complete; MEDLINE; PsycINFO, Web of Science and Scopus with a protocol registered with Open Science Framework DOI 10.17605/OSF.IO/5SRMV. RESULTS After screening 1145 papers there were 7 paper that met the inclusion criteria. Statistically significant associations were found in five of the studies which identify higher abundance of potentially pathogenic bacteria such as Erwinia, Serratia, T mayombie, Bacteroides with higher maternal stress, and lower levels of stress linked to potentially beneficial bacteria such Lactococcus, Lactobacillus, Akkermansia. However, one study presents conflicting results where it was reported that higher maternal stress was linked to the prevalence of more beneficial bacteria. CONCLUSION This review suggests that maternal stress does have an impact on the alteration of abundance and diversity of influential bacteria in the gut microbiome, however, it can affect colonisation in different ways. These bacterial changes have the capacity to influence long term health and disease. The review analyses data collection tools and methods, offers potential reasons for these findings as well as suggestions for future research.
Collapse
Affiliation(s)
- Niamh Ryan
- School of Nursing and Midwifery, University College Cork, Cork, Ireland
| | - Siobhain O’Mahony
- Department of Anatomy and Neuroscience, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Lloyd Philpott
- School of Nursing and Midwifery, University College Cork, Cork, Ireland
| | - Helen Mulcahy
- School of Nursing and Midwifery, University College Cork, Cork, Ireland
| |
Collapse
|
4
|
Malham M, Vestergaard MV, Bataillon T, Villesen P, Dempfle A, Bang C, Engsbro AL, Jakobsen C, Franke A, Wewer V, Thingholm LB, Petersen AM. The Composition of the Fecal and Mucosa-adherent Microbiota Varies Based on Age and Disease Activity in Ulcerative Colitis. Inflamm Bowel Dis 2025; 31:501-513. [PMID: 39150994 DOI: 10.1093/ibd/izae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Indexed: 08/18/2024]
Abstract
BACKGROUND Pediatric-onset ulcerative colitis (pUC) represents a more aggressive disease phenotype compared with adult-onset UC. We hypothesized that this difference can, in part, be explained by the composition of the microbiota. METHODS In a prospective, longitudinal study, we included pediatric (N = 30) and adult (N = 30) patients with newly or previously (>1 year) diagnosed UC. We analyzed the microbiota composition in the mucosa-adherent microbiota at baseline, using 16S rRNA gene sequencing, and the fecal microbiota at baseline and at 3-month intervals, using shotgun metagenomics. RESULTS For fecal samples, the bacterial composition differed between pUC and aUC in newly diagnosed patients (β-diversity, Bray Curtis: R2 = 0.08, P = .02). In colon biopsies, microbial diversity was higher in aUC compared with pUC (α-diversity, Shannon: estimated difference 0.54, P = .006). In the mucosa-adherent microbiota, Alistipes finegoldii was negatively associated with disease activity in pUC while being positively associated in aUC (estimate: -0.255 and 0.098, P = .003 and P = .02 in pUC and aUC, respectively). Finally, we showed reduced stability of the fecal microbiota in pediatric patients, evidenced by a different composition of the fecal microbiota in newly and previously diagnosed pUC, a pattern not found in adults. CONCLUSIONS Our results indicate that pediatric UC patients have a more unstable fecal microbiota and a lower α diversity than adult patients and that the microbiota composition differs between aUC and pUC patients. These findings offer some explanation for the observed differences between pUC and aUC and indicate that individualized approaches are needed if microbiota modifications are to be used in the future treatment of UC.
Collapse
Affiliation(s)
- Mikkel Malham
- Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents, and Adults, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - Marie V Vestergaard
- Bioinformatics Research Centre, Aarhus University, 8000 Aarhus C, Denmark
- Center for Molecular Prediction of Inflammatory Bowel Disease, PREDICT, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
| | - Thomas Bataillon
- Bioinformatics Research Centre, Aarhus University, 8000 Aarhus C, Denmark
| | - Palle Villesen
- Bioinformatics Research Centre, Aarhus University, 8000 Aarhus C, Denmark
- Department of Clinical Medicine, Aarhus University Hospital, 8000 Aarhus C, Denmark
| | - Astrid Dempfle
- Institut für Medizinische Informatik und Statistik, Universitätsklinikum Schleswig-Holstein, 24105 Kiel, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany
| | - Anne Line Engsbro
- Department of Clinical Microbiology, Copenhagen University Hospital-Hvidovre, Denmark
| | - Christian Jakobsen
- Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents, and Adults, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany
| | - Vibeke Wewer
- Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents, and Adults, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - Louise B Thingholm
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany
| | - Andreas M Petersen
- Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital-Hvidovre, Denmark
- Gastrounit, Medical Division, Copenhagen University Hospital-Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Bakshani CR, Ojuri TO, Pilgaard B, Holck J, McInnes R, Kozak RP, Zakhour M, Çakaj S, Kerouedan M, Newton E, Bolam DN, Crouch LI. Carbohydrate-active enzymes from Akkermansia muciniphila break down mucin O-glycans to completion. Nat Microbiol 2025; 10:585-598. [PMID: 39891011 PMCID: PMC11790493 DOI: 10.1038/s41564-024-01911-7] [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: 04/23/2024] [Accepted: 12/10/2024] [Indexed: 02/03/2025]
Abstract
Akkermansia muciniphila is a human microbial symbiont residing in the mucosal layer of the large intestine. Its main carbon source is the highly heterogeneous mucin glycoprotein, and it uses an array of carbohydrate-active enzymes and sulfatases to access this complex energy source. Here we describe the biochemical characterization of 54 glycoside hydrolases, 11 sulfatases and 1 polysaccharide lyase from A. muciniphila to provide a holistic understanding of their carbohydrate-degrading activities. This was achieved using a variety of liquid chromatography techniques, mass spectrometry, enzyme kinetics and thin-layer chromatography. These results are supported with A. muciniphila growth and whole-cell assays. We find that these enzymes can act synergistically to degrade the O-glycans on the mucin polypeptide to completion, down to the core N-acetylgalactosaime. In addition, these enzymes can break down human breast milk oligosaccharide, ganglioside and globoside glycan structures, showing their capacity to target a variety of host glycans. These data provide a resource to understand the full degradative capability of the gut microbiome member A. muciniphila.
Collapse
Affiliation(s)
- Cassie R Bakshani
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Taiwo O Ojuri
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Bo Pilgaard
- Protein Chemistry and Enzyme Technology Section, DTU Bioengineering, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Jesper Holck
- Protein Chemistry and Enzyme Technology Section, DTU Bioengineering, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Ross McInnes
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | | | - Maria Zakhour
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Sara Çakaj
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Manon Kerouedan
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Emily Newton
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - David N Bolam
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Lucy I Crouch
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
6
|
Zhao C, Li Y, Wang H, Solomon AI, Wang S, Dong X, Song B, Ren Z. Dietary supplementation with compound microecological preparations: effects on the production performance and gut microbiota of lactating female rabbits and their litters. Microbiol Spectr 2025; 13:e0006724. [PMID: 39611688 PMCID: PMC11705915 DOI: 10.1128/spectrum.00067-24] [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: 01/06/2024] [Accepted: 08/04/2024] [Indexed: 11/30/2024] Open
Abstract
Early weaning is frequently accompanied by a significant increase in diarrhea and mortality rates, which reduces rabbits' performance. Although antibiotics can reduce pathogenic bacteria, they also harm beneficial microorganisms and disrupt the normal intestinal microbiota balance. In order to find non-residue and non-toxic alternatives to antibiotics to ensure the safety of animal products, we conducted a study on the effect of compound microecological preparations supplementation on lactating female rabbits and their offspring. A total of 60 female rabbits were randomly assigned to four groups: CON, supplemented with probiotics at 3, 6, and 9 g/female rabbit/day from day 24 of gestation until weaning. We observed that probiotics supplementation significantly enhanced production performance (P < 0.05), immune and antioxidant function (P < 0.05), as well as intestinal flora composition in lactating rabbits and their offspring. Notably, compared with the control group, the experimental group exhibited a 19.23%, 44.22%, and 24.57% increase in milk yield (P = 0.002). Regarding rabbit growth performance, the average body weight of young rabbits in the experimental group showed a significant increase of 3.59%, 10.22%, and 6.74% at day 35 (P = 0.022), whereas the average daily gain (ADG) of rabbits aged between 21 and 35 days was significantly elevated by 4.94%, 17.06%, and 6.28% in the experimental group (P < 0.001). In conclusion, probiotics supplementation can significantly enhance lactation performance, promote growth and disease resistance in rabbits, as well as improve intestinal health when administered at a dosage of 6 g/day. Moreover, the limited sample size in this study may hinder the detection of subtle effects, and augmenting the sample size will bolster the reliability of the study findings. IMPORTANCE The intestinal environment of rabbits is fragile and susceptible to environmental influences, leading to inflammatory intestinal diseases. Adding antibiotics to rabbit feed can achieve the effect of preventing and treating inflammation, which can also lead to the imbalance of the gut microbiota and residual antibiotics in agricultural products. Composite probiotics are live microbial feed additives composed of various ratios of probiotics and have become the most promising alternative to antibiotics due to their residue-free and non-toxic properties. The aim of this study was to investigate the impact of compound probiotics on lactating female rabbits and their offspring. Our findings highlight the potential of compound microecological preparations as an effective strategy for enhancing lactation performance, immune function, and antioxidant capacity in rabbits. The supplementation of probiotics through rabbit milk offers a promising approach to optimize the growth and health outcomes of newborn rabbits.
Collapse
Affiliation(s)
- Chengcheng Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Youhao Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Hui Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Ahamba Ifeanyi Solomon
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Shuhui Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xianggui Dong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Bing Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhanjun Ren
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
7
|
Liu H, Huang R, Shen B, Huang C, Zhou Q, Xu J, Chen S, Lin X, Wang J, Zhao X, Guo Y, Ai X, Liu Y, Wang Y, Zhang W, Zhi F. Live Akkermansia muciniphila boosts dendritic cell retinoic acid synthesis to modulate IL-22 activity and mitigate colitis in mice. MICROBIOME 2024; 12:275. [PMID: 39734222 PMCID: PMC11684322 DOI: 10.1186/s40168-024-01995-7] [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: 05/10/2024] [Accepted: 12/02/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND The interplay between gut microbiota and immune responses is crucial in ulcerative colitis (UC). Though Akkermansia muciniphila (Akk) shows therapeutic potential, the mechanisms remain unclear. This study sought to investigate differences in therapeutic efficacy among different forms or strains of Akk and elucidate the underlying mechanisms. RESULTS Employing a dextran sulfate sodium (DSS)-induced colitis mouse model, we assessed Akk's impact on colitis using cellular cytokine analysis, immune phenotyping, proteomics, and biochemical methods. Our results suggest that treatment with live Akk effectively reduced colitis in the DSS-induced model, whereas heat-inactivated Akk did not yield the same results. Notably, Akk exhibited protective properties by promoting the secretion of IL-22 by Group 3 innate lymphoid cells (ILC3s), as evidenced by the absence of protection in IL-22 knockout mice. Additionally, Akk augmented the population of CD103+CD11b- dendritic cells (DCs) and enhanced their retinoic acid (RA) synthesis through the modulation of RALDH2, a crucial enzyme in RA metabolism. The depletion of RALDH2 in DCs diminished Akk's protective properties and impaired IL-22-mediated mucosal healing. Mechanistically, Akk activated RA production in DCs by enhancing the JAK2-STAT3 signaling pathway. Additionally, various strains of Akk may exhibit differing abilities to alleviate colitis, with the novel strain Am06 derived from breast milk showing consistent efficacy similar to the reference strain. CONCLUSIONS In summary, our findings indicate that certain strains of Akk may mitigate colitis through the promotion of RA synthesis and IL-22 secretion, underscoring the potential efficacy of Akk as a therapeutic intervention for the management of UC. Video Abstract.
Collapse
Affiliation(s)
- Hongbin Liu
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruo Huang
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Binhai Shen
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chongyang Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qian Zhou
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiahui Xu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shengbo Chen
- Department of Gastroenterology, Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Xinlong Lin
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinmei Zhao
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yandong Guo
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiuyun Ai
- Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yangyang Liu
- Guangzhou ZhiYi Biotechnology Co., Ltd, Guangzhou, China
| | - Ye Wang
- Guangzhou ZhiYi Biotechnology Co., Ltd, Guangzhou, China
| | - Wendi Zhang
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Fachao Zhi
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Shen XH, Guan J, Lu DP, Hong SC, Yu L, Chen X. Peptostreptococcus Anaerobius enhances dextran sulfate sodium-induced colitis by promoting nf-κB-NLRP3-Dependent macrophage pyroptosis. Virulence 2024; 15:2435391. [PMID: 39611567 PMCID: PMC11610558 DOI: 10.1080/21505594.2024.2435391] [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: 11/17/2023] [Revised: 09/22/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024] Open
Abstract
Evidence indicates that gut microbiota is crucial in ulcerative colitis (UC) development. Increased Peptostreptococcus species abundance is linked to UC, but its role and mechanisms in intestinal inflammation are not well understood. This study used a dextran sulfate sodium (DSS)-induced colitis model in mice, and different bacterial strains were administered via gavage. We assessed clinical manifestations, colonic barrier function, gut microbiota composition, and levels of inflammatory cytokines, NOD-like receptor family pyrin domain-containing 3 (NLRP3) signaling molecules, and pyroptosis-related proteins. Mouse bone marrow-derived macrophages (BMDMs) were infected with Peptostreptococcus anaerobius at different time points and multiplicities of infection (MOI). Cell viability and the expression of NLRP3 signaling molecules and pyroptosis-associated proteins were assessed. The inhibitors C29, TAK-242, and MCC950 were employed for Toll-like receptor (TLR) and NLRP3 signaling pathways. It was observed that P. anaerobius exacerbated intestinal inflammation and barrier injury in DSS-induced colitis in mice. Additionally, P. anaerobius contributed to gut microbiota dysbiosis during colitis progression. P. anaerobius induced the expression of NLRP3 signaling molecules and pyroptosis-associated proteins in mouse colitis tissues. In vitro assays demonstrated that P. anaerobius activated NLRP3 inflammasome and evoked gasdermin D-mediated pyroptosis and interleukin (IL)-1β secretion in macrophages. Furthermore, TLR2 and TLR4 were identified as key mediators of P. anaerobius-induced macrophage pyroptosis via activation of the Nuclear Factor-kappa B (NF-κB)-NLRP3 pathway. In conclusion, P. anaerobius promotes macrophage pyroptosis and IL-1β secretion through the TLR2/4-NF-κB-NLRP3 signaling axis, thereby aggravating colitis. P. anaerobius may represent a potential risk factor for UC development.
Collapse
Affiliation(s)
- Xu-Hang Shen
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory of Digestive Disease, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Guan
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory of Digestive Disease, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - De-Peng Lu
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory of Digestive Disease, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shao-Cheng Hong
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory of Digestive Disease, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Yu
- Anhui Provincial Key Laboratory of Zoonoses, Anhui Medical University, Hefei, China
- Department of Microbiology and Parasitology, Anhui Medical University, Hefei, China
| | - Xi Chen
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory of Digestive Disease, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
9
|
Hertz S, Anderson JM, Nielsen HL, Schachtschneider C, McCauley KE, Özçam M, Larsen L, Lynch SV, Nielsen H. Fecal microbiota is associated with extraintestinal manifestations in inflammatory bowel disease. Ann Med 2024; 56:2338244. [PMID: 38648495 PMCID: PMC11036898 DOI: 10.1080/07853890.2024.2338244] [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: 09/29/2023] [Revised: 02/15/2024] [Accepted: 03/17/2024] [Indexed: 04/25/2024] Open
Abstract
INTRODUCTION A large proportion of patients with inflammatory bowel disease (IBD) experience IBD-related inflammatory conditions outside of the gastrointestinal tract, termed extraintestinal manifestations (EIMs) which further decreases quality of life and, in extreme cases, can be life threatening. The pathogenesis of EIMs remains unknown, and although gut microbiota alterations are a well-known characteristic of patients with IBD, its relationship with EIMs remains sparsely investigated. This study aimed to compare the gut microbiota of patients with IBD with and without EIMs. METHODS A total of 131 Danish patients with IBD were included in the study, of whom 86 had a history of EIMs (IBD-EIM) and 45 did not (IBD-C). Stool samples underwent 16S rRNA sequencing. Amplicon sequence variants (ASVs) were mapped to the Silva database. Diversity indices and distance matrices were compared between IBD-EIM and IBD-C. Differentially abundant ASVs were identified using a custom multiple model statistical analysis approach, and modules of co-associated bacteria were identified using sparse correlations for compositional data (SparCC) and related to patient EIM status. RESULTS Patients with IBD and EIMs exhibited increased disease activity, body mass index, increased fecal calprotectin levels and circulating monocytes and neutrophils. Microbiologically, IBD-EIM exhibited lower fecal microbial diversity than IBD-C (Mann-Whitney's test, p = .01) and distinct fecal microbiota composition (permutational multivariate analysis of variance; weighted UniFrac, R2 = 0.018, p = .01). A total of 26 ASVs exhibited differential relative abundances between IBD-EIM and IBD-C, including decreased Agathobacter and Blautia and increased Eggerthella lenta in the IBD-EIM group. SparCC analysis identified 27 bacterial co-association modules, three of which were negatively related to EIM (logistic regression, p < .05) and included important health-associated bacteria, such as Agathobacter and Faecalibacterium. CONCLUSIONS The fecal microbiota in IBD patients with EIMs is distinct from that in IBD patients without EIM and could be important for EIM pathogenesis.
Collapse
Affiliation(s)
- Sandra Hertz
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Jacqueline Moltzau Anderson
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Hans Linde Nielsen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Clinical Microbiology, Aalborg University Hospital, Aalborg, Denmark
| | - Claire Schachtschneider
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Kathryn E. McCauley
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Mustafa Özçam
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Lone Larsen
- Department of Gastroenterology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Center for Molecular Prediction of Inflammatory Bowel Disease, PREDICT, Aalborg University, Aalborg, Denmark
| | - Susan V. Lynch
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Henrik Nielsen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
10
|
He X, Zhang Y. Changes in gut flora in patients with epilepsy: a systematic review and meta-analysis. Front Microbiol 2024; 15:1480022. [PMID: 39611090 PMCID: PMC11602489 DOI: 10.3389/fmicb.2024.1480022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024] Open
Abstract
Background Epilepsy is a prevalent chronic neurological disorder that is strongly associated with a wide range of psychological, cognitive and social problems. It affects a significant proportion of the global population and has a number of complex etiologies. A growing body of research indicates that there is a strong association between epilepsy and the gut microbiota. Indeed, a substantial body of research has investigated the potential role of epilepsy in relation to the gut microbiota, examining alterations in the abundance, diversity, and relative abundance of the gut microbiota in patients with epilepsy. Methods This study was conducted in accordance with the PRISMA guidelines and included multiple studies that met specific criteria. A keyword search was conducted in the following databases: PubMed, Embase, and Web of Science. The data extraction and quality assessment were conducted by two independent researchers. A systematic review and meta-analysis of the relationship between patients with epilepsy and gut flora was conducted using the R 4.3.4 software. Results The results of the analyses indicated that the intestinal flora of patients with epilepsy did not differ significantly in alpha diversity compared to healthy controls. However, the relative abundance of specific flora, such as Verrucomicrobia and Ackermannia was significantly increased in patients, whereas Lactobacillus was significantly decreased. Conclusion The relationship between epilepsy and gut flora is reciprocal. The present meta-analysis demonstrated that there were no statistically significant alterations in the overall characteristics of the intestinal flora of the patients. However, significant changes were observed in the relative abundance of certain phyla and genera. Consequently, it is hypothesized that epilepsy can cause changes in the relative abundance of specific flora in patients. Furthermore, in conjunction with previous studies, it is believed that changes in intestinal flora can also have an effect on seizures. For example, Lactobacillus may be a beneficial genus that potentially reduces seizures. Conversely, the effect of Akkermansia is two-sided.
Collapse
Affiliation(s)
- Xingyan He
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Yuxin Zhang
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
Chen WX, Yan QX, Zhong RZ, Tang SX, Loor JJ, Tan ZL. A type 2 immune circuit and arachidonic acid metabolism role in anti-nematode infection: evidence from transcriptome and targeted metabolome data in goat. Animal 2024; 18:101338. [PMID: 39405961 DOI: 10.1016/j.animal.2024.101338] [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: 12/27/2023] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 11/18/2024] Open
Abstract
The gastrointestinal nematode infection poses a covert threat to both humans and domestic animals worldwide, eliciting a type 2 immune response within the small intestine. Intestinal tuft cells detect the nematode and activated group 2 innate lymphoid cells. Tuft cell-derived leukotrienes (one of the metabolites of arachidonic acid) were found to drive rapid anti-helminth immunity, but it is still poorly understood whether the tuft cell-mediated type 2 immune circuit and arachidonic acid metabolism modulate anti-parasitic immunity in the gastric epithelium. This study was designed to evaluate the immunological responses of goats inoculated with or without H. contortus. Results showed that H. contortus infection induced a systemic type 2 immune response, characterised by lymphocyte proliferation and greater eosinophils both in peripheral blood and abomasal mucosa, as well as increased type 2 cytokines IL-4, IL-5, and IL-13. Infection of H. contortus altered the transcriptome of the abomasum epithelium, especially tuft cell-mediated circuit-key genes. The infection also influenced the abomasal microbiota, arachidonic acid metabolism and related lipid metabolites, accompanying with great increases in the secretion of leukotrienes and prostaglandins. These findings demonstrate the role of tuft cells mediated circuit in sensing H. contortus infection and immune activation, reveal the candidate function of arachidonic acid involved in anti-helminth immunity, and suggest novel strategies for the control of parasitic diseases in livestock and humans.
Collapse
Affiliation(s)
- W X Chen
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, PR China; Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
| | - Q X Yan
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, PR China.
| | - R Z Zhong
- Jilin Provincial Key Laboratory of Grassland Farming, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun 130102, Jilin, PR China
| | - S X Tang
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, PR China
| | - J J Loor
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Z L Tan
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, PR China
| |
Collapse
|
12
|
Luo C, Du Y, Zhu R, Qi Q, Luo S, Feng X. Effects of Akkermansia muciniphila on Gut Morphology, Antioxidant Indices, and Gut Microbiome of Mice Under Heat Stress. Foodborne Pathog Dis 2024; 21:724-730. [PMID: 39082080 DOI: 10.1089/fpd.2024.0046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2024] Open
Abstract
Nutritional manipulations can reduce the detrimental effects of heat stress on animal health and production. Akkermansia muciniphila (AM) is an innovative beneficial bacteria and can be used for conventional use as dietary supplements and pharmaceutical application. This study aimed to investigate the effects of administering AM on gut morphology, antioxidant indices, and gut microbiome of mice during heat stress. A total of 24 BALB/c mice were randomly assigned to three groups including the control group (CON), heat stress group (HS), and AM administration under heat stress group (AM). Our results showed heat stress significantly increased the water consumption of mice. Administration of AM did not improve feed intake or weight gain. The serum levels of alanine aminotransferase and aspartate aminotransferase as well as antioxidant parameters were not different among the three groups. Heat stress decreased the jejunal villus height, and AM could reverse this effect. AM administration significantly increased the relative abundance of Verrucomicrobiota at the phylum level. At the genus level, heat stress and AM groups tended to have a lower abundance of Alloprevotella. In addition, AM tended to increase the relative abundance of [Eubacterium]_xylanophilum_group in comparison with the other two groups. In summary, administration of AM can alleviate the damage of heat stress to the jejunum. However, it has no effect on serum antioxidant parameters, and its effect on the cecal microbiota is limited.
Collapse
Affiliation(s)
- Caiyu Luo
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yingzhu Du
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Rongxia Zhu
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Qien Qi
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Shumeng Luo
- The Hong Kong Polytechnic University, Hong Kong, China
| | - Xin Feng
- School of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|
13
|
Dai N, Yang X, Pan P, Zhang G, Sheng K, Wang J, Liang X, Wang Y. Bacillus paralicheniformis, an acetate-producing probiotic, alleviates ulcerative colitis via protecting the intestinal barrier and regulating the NLRP3 inflammasome. Microbiol Res 2024; 287:127856. [PMID: 39079268 DOI: 10.1016/j.micres.2024.127856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 08/22/2024]
Abstract
Ulcerative colitis (UC) presents a challenging scenario in digestive health, characterized by recurrent inflammation that is often hard to manage. Bacteria capable of producing short-chain fatty acids (SCFAs) play a pivotal role in mitigating UC symptoms, rendering them promising candidates for probiotic therapy. In this investigation, we assessed the impact of Bacillus paralicheniformis HMPM220325 on dextran sodium sulfate (DSS)-induced UC in mice. Genomic analysis of the strain revealed the presence of protease genes associated with acetate and butyrate synthesis, with acetic acid detected in its fermentation broth. Administration of B. paralicheniformis HMPM220325 to UC mice ameliorated pathological manifestations of the condition and restored intestinal barrier function. Furthermore, B. paralicheniformis HMPM220325 suppressed the activation of the NLRP3 inflammasome signaling pathway and modulated the composition of the intestinal microbiota. These findings shed significant light on the potential of B. paralicheniformis as a probiotic candidate, offering a novel avenue for the prevention and therapeutic intervention of colitis.
Collapse
Affiliation(s)
- Nini Dai
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Xinting Yang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Peilong Pan
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Guanghui Zhang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Kangliang Sheng
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Jingmin Wang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Xiao Liang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China.
| | - Yongzhong Wang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China.
| |
Collapse
|
14
|
Etlin S, Rose J, Bielski L, Walter C, Kleinman AS, Mason CE. The human microbiome in space: parallels between Earth-based dysbiosis, implications for long-duration spaceflight, and possible mitigation strategies. Clin Microbiol Rev 2024; 37:e0016322. [PMID: 39136453 PMCID: PMC11391694 DOI: 10.1128/cmr.00163-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
SUMMARYThe human microbiota encompasses the diverse communities of microorganisms that reside in, on, and around various parts of the human body, such as the skin, nasal passages, and gastrointestinal tract. Although research is ongoing, it is well established that the microbiota exert a substantial influence on the body through the production and modification of metabolites and small molecules. Disruptions in the composition of the microbiota-dysbiosis-have also been linked to various negative health outcomes. As humans embark upon longer-duration space missions, it is important to understand how the conditions of space travel impact the microbiota and, consequently, astronaut health. This article will first characterize the main taxa of the human gut microbiota and their associated metabolites, before discussing potential dysbiosis and negative health consequences. It will also detail the microbial changes observed in astronauts during spaceflight, focusing on gut microbiota composition and pathogenic virulence and survival. Analysis will then turn to how astronaut health may be protected from adverse microbial changes via diet, exercise, and antibiotics before concluding with a discussion of the microbiota of spacecraft and microbial culturing methods in space. The implications of this review are critical, particularly with NASA's ongoing implementation of the Moon to Mars Architecture, which will include weeks or months of living in space and new habitats.
Collapse
Affiliation(s)
- Sofia Etlin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- Department of Biology, Cornell University, Ithaca, New York, USA
- BioAstra Inc., New York, New York, USA
| | - Julianna Rose
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- Department of Biology, Cornell University, Ithaca, New York, USA
- BioAstra Inc., New York, New York, USA
| | - Luca Bielski
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- Department of Biology, Cornell University, Ithaca, New York, USA
| | - Claire Walter
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- Department of Biology, Cornell University, Ithaca, New York, USA
- BioAstra Inc., New York, New York, USA
| | - Ashley S Kleinman
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- BioAstra Inc., New York, New York, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York, USA
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
- Tri-Institutional Biology and Medicine program, Weill Cornell Medicine, New York, New York, USA
- WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
15
|
Pusa T, Rousu J. Stable biomarker discovery in multi-omics data via canonical correlation analysis. PLoS One 2024; 19:e0309921. [PMID: 39250478 PMCID: PMC11383239 DOI: 10.1371/journal.pone.0309921] [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: 02/27/2024] [Accepted: 08/20/2024] [Indexed: 09/11/2024] Open
Abstract
Multi-omics analysis offers a promising avenue to a better understanding of complex biological phenomena. In particular, untangling the pathophysiology of multifactorial health conditions such as the inflammatory bowel disease (IBD) could benefit from simultaneous consideration of several omics levels. However, taking full advantage of multi-omics data requires the adoption of suitable new tools. Multi-view learning, a machine learning technique that natively joins together heterogeneous data, is a natural source for such methods. Here we present a new approach to variable selection in unsupervised multi-view learning by applying stability selection to canonical correlation analysis (CCA). We apply our method, StabilityCCA, to simulated and real multi-omics data, and demonstrate its ability to find relevant variables and improve the stability of variable selection. In a case study on an IBD microbiome data set, we link together metagenomics and metabolomics, revealing a connection between their joint structure and the disease, and identifying potential biomarkers. Our results showcase the usefulness of multi-view learning in multi-omics analysis and demonstrate StabilityCCA as a powerful tool for biomarker discovery.
Collapse
Affiliation(s)
- Taneli Pusa
- Department of Computer Science, Aalto University, Espoo, Finland
| | - Juho Rousu
- Department of Computer Science, Aalto University, Espoo, Finland
| |
Collapse
|
16
|
Huang J, Zhang J, Wang F, Tang X. Modified Gegen Qinlian Decoction modulated the gut microbiome and bile acid metabolism and restored the function of goblet cells in a mouse model of ulcerative colitis. Front Immunol 2024; 15:1445838. [PMID: 39165355 PMCID: PMC11333261 DOI: 10.3389/fimmu.2024.1445838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 07/15/2024] [Indexed: 08/22/2024] Open
Abstract
Objective Modified Gegen Qinlian Decoction (MGQD) has been shown to effectively relieve ulcerative colitis (UC) without a known pharmacological mechanism. In this study, the anti-colitis efficaciousness of MGQD and its underlying mechanisms in UC were evaluated. Methods Mice with colitis were administered MGQD for 7 days. Following the evaluation of clinical symptoms, gut microbiota in the feces of UC mice was examined using 16S rRNA sequencing and bile acids (BAs) were examined using LC/MS. Gut microbiota consumption and fecal microbiota transplantation (FMT) were used to explore the involvement of gut microbiota in the anti-UC action of MGQD. Results MGQD relieved colitis as shown by weight loss protection, a lower disease activity index (DAI), restoration of intestinal length reduction, and lower histopathologic scores. MGQD also restored crypt stem cell proliferation and function of colonic goblet cells, and promoted MUC2 protein secretion. Interestingly, investigations using gut bacterial depletion and FMT showed that MGQD attenuated colonic damage in a gut-dependent way. The modulation of the gut microbiota by MGQD might be attributed to a decrease in Odoribacter and an increase in norank_f_Muribaculaceae. In addition, MGQD modulated the metabolism of BAs while restoring the structure of the gut microbiota. Conclusion MGQD significantly alleviated colitis in mice, which may be associated with the modulation of gut microbiota and BA metabolism and restoration of function of goblet cells. However, factors other than the gut microbiota may also be involved in the amelioration of UC by MGQD.
Collapse
Affiliation(s)
- Jinke Huang
- Department of Gastroenterology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiaqi Zhang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengyun Wang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Xudong Tang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
17
|
Mo C, Lou X, Xue J, Shi Z, Zhao Y, Wang F, Chen G. The influence of Akkermansia muciniphila on intestinal barrier function. Gut Pathog 2024; 16:41. [PMID: 39097746 PMCID: PMC11297771 DOI: 10.1186/s13099-024-00635-7] [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: 06/09/2024] [Accepted: 07/20/2024] [Indexed: 08/05/2024] Open
Abstract
Intestinal barriers play a crucial role in human physiology, both in homeostatic and pathological conditions. Disruption of the intestinal barrier is a significant factor in the pathogenesis of gastrointestinal inflammatory diseases, such as inflammatory bowel disease. The profound influence of the gut microbiota on intestinal diseases has sparked considerable interest in manipulating it through dietary interventions, probiotics, and fecal microbiota transplantation as potential approaches to enhance the integrity of the intestinal barrier. Numerous studies have underscored the protective effects of specific microbiota and their associated metabolites. In recent years, an increasing body of research has demonstrated that Akkermansia muciniphila (A. muciniphila, Am) plays a beneficial role in various diseases, including diabetes, obesity, aging, cancer, and metabolic syndrome. It is gaining popularity as a regulator that influences the intestinal flora and intestinal barrier and is recognized as a 'new generation of probiotics'. Consequently, it may represent a potential target and promising therapy option for intestinal diseases. This article systematically summarizes the role of Am in the gut. Specifically, we carefully discuss key scientific issues that need resolution in the future regarding beneficial bacteria represented by Am, which may provide insights for the application of drugs targeting Am in clinical treatment.
Collapse
Affiliation(s)
- Chunyan Mo
- Medical School, Kunming University of Science and Technology, 727 Jingming South Road, Chenggong District, Kunming, 650500, China
| | - Xiran Lou
- Medical School, Kunming University of Science and Technology, 727 Jingming South Road, Chenggong District, Kunming, 650500, China
| | - Jinfang Xue
- Medical School, Kunming University of Science and Technology, 727 Jingming South Road, Chenggong District, Kunming, 650500, China
| | - Zhuange Shi
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650034, China
| | - Yifang Zhao
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650034, China
| | - Fuping Wang
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650034, China
| | - Guobing Chen
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650034, China.
| |
Collapse
|
18
|
Chen J, Xu W, Liu Y, Liang X, Chen Y, Liang J, Cao J, Lu B, Sun C, Wang Y. Lonicera japonica Thunb. and its characteristic component chlorogenic acid alleviated experimental colitis by promoting Lactobacillus and fecal short‐chain fatty acids production. FOOD FRONTIERS 2024; 5:1583-1602. [DOI: 10.1002/fft2.412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024] Open
Abstract
AbstractUlcerative colitis is intricately linked to intestinal oxidative stress and dysbiosis of the gut microbiota. Lonicera japonica Thunb. (LJ) is a traditional edible and medical flower in China, and chlorogenic acid (CGA) is one of its characteristic components. However, it remains unclear whether gut microbiota plays a role in the therapeutic effects of LJ and GCA on colitis. Here, we first observed that oral administration of LJ and CGA for 3 weeks dramatically promoted the growth of Lactobacillus and fecal short‐chain fatty acids (SCFAs) production in healthy mice. Subsequently, the alleviating effects of LJ and CGA on colitis were explored with a dextran sulfate sodium‐induced colitis mice model. The intervention of LJ and CGA notably alleviated inflammation, intestinal barrier impairment, and oxidative stress in colitis and led to a significant elevation in Lactobacillus and fecal SCFAs. Eventually, the key role of gut microbiota and their metabolites on the therapeutic effects was validated by performing fecal microbiota transplantation and sterile fecal suspensions transplantation from LJ and CGA‐treated healthy mice to colitis mice. Our findings demonstrated that consumption of LJ and CGA could benefit the host both in healthy condition and colitis. The beneficial effects were attributed to the improvement of the endogenous antioxidant system and promotion of the probiotic Lactobacillus and SCFAs production. Our study highlighted the great potential of LJ and CGA to be consumed as functional foods and provided novel mechanisms by which they alleviated colitis.
Collapse
Affiliation(s)
- Jiebiao Chen
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement Zhejiang University, Zijingang Campus Hangzhou People's Republic of China
| | - Wanhua Xu
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement Zhejiang University, Zijingang Campus Hangzhou People's Republic of China
| | - Yang Liu
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement Zhejiang University, Zijingang Campus Hangzhou People's Republic of China
- Shandong (Linyi) Institute of Modern Agriculture Zhejiang University Linyi Shandong People's Republic of China
| | - Xiao Liang
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement Zhejiang University, Zijingang Campus Hangzhou People's Republic of China
| | - Yunyi Chen
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement Zhejiang University, Zijingang Campus Hangzhou People's Republic of China
| | - Jiaojiao Liang
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement Zhejiang University, Zijingang Campus Hangzhou People's Republic of China
| | - Jinping Cao
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement Zhejiang University, Zijingang Campus Hangzhou People's Republic of China
- Hainan Institute of Zhejiang University, Zhejiang University Sanya Hainan People's Republic of China
| | - Baiyi Lu
- College of Biosystems Engineering and Food Science, Key Laboratory for Quality Evaluation and Health Benefit of Agro‐Products of Ministry of Agriculture and Rural Affairs, Key Laboratory for Quality and Safety Risk Assessment of Agro‐Products Storage and Preservation of Ministry of Agriculture and Rural Affairs Zhejiang University Hangzhou People's Republic of China
| | - Chongde Sun
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement Zhejiang University, Zijingang Campus Hangzhou People's Republic of China
- Hainan Institute of Zhejiang University, Zhejiang University Sanya Hainan People's Republic of China
| | - Yue Wang
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement Zhejiang University, Zijingang Campus Hangzhou People's Republic of China
- Shandong (Linyi) Institute of Modern Agriculture Zhejiang University Linyi Shandong People's Republic of China
| |
Collapse
|
19
|
Li N, Yuan Q, Qi Y, Wu P, Cui S, Zheng G. The Potential Implications of Sex-Specific Differences in the Intestinal Bacteria of the Overwintering Wolf Spider Pardosa astrigera (Araneae: Lycosidae). INSECTS 2024; 15:490. [PMID: 39057223 PMCID: PMC11276740 DOI: 10.3390/insects15070490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024]
Abstract
Gut microbiota can promote the resistance of host arthropods to low-temperature stress. Female Pardosa astrigera have a lower anti-freeze compound level and weaker resistance to cold temperatures than the males in winter, which implies that their intestinal bacteria may be different during overwintering. This study primarily compared the intestinal bacterial communities between the two sexes of P. astrigera in a temperate region using 16S rRNA gene sequencing. Our findings indicated that the Chao1 and Shannon indices of intestinal bacteria in females were significantly higher than those in males, while the Simpson index in females was significantly lower than that in males. The male intestinal bacterial community was characterized by Proteobacteria and Actinobacteriota at the phylum level and by Pseudomonas and Rhodococcus at the genus level, with total relative abundances of 89.58% and 85.22%, respectively, which were also significantly higher than those in females, whose total relative abundances were 47.49% and 43.68%, respectively. In contrast, the total relative abundances of Bacteroidota and Firmicutes were significantly lower in males (4.26% and 4.75%, respectively) than in females (26.25% and 22.31%, respectively). Noteworthy divergences in bacterial communities were also found through an LEfSe analysis between females and males. Additionally, the results of the PICRUSt2 analysis showed that six out of eleven level-2 pathways related to key metabolic functions were significantly (or marginally significantly) higher in females than males, and five other level-2 pathways were significantly (or marginally significantly) lower in females than males. Our results imply that significant gender differences exist in intestinal bacterial communities of overwintering P. astrigera. We suggest that Pseudomonas versuta (belonging to Proteobacteria) and Rhodococcus erythropolis (belonging to Actinobacteriota) may have the potential to play key roles in overwintering P. astrigera.
Collapse
Affiliation(s)
- Ningkun Li
- College of Life Science, Shenyang Normal University, Shenyang 110034, China
| | - Quan Yuan
- College of Life Science, Shenyang Normal University, Shenyang 110034, China
| | - Yaru Qi
- College of Life Science, Shenyang Normal University, Shenyang 110034, China
| | - Pengfeng Wu
- College of Life Science, Shenyang Normal University, Shenyang 110034, China
| | - Shuyan Cui
- College of Life Science, Shenyang Normal University, Shenyang 110034, China
| | - Guo Zheng
- College of Life Science, Shenyang Normal University, Shenyang 110034, China
- Liaoning Key Laboratory for Biological Evolution and Agricultural Ecology, Shenyang 110034, China
| |
Collapse
|
20
|
Filippo D, Guardone L, Listorti V, Elisabetta R. Microbiome in cancer: A comparative analysis between humans and dogs. Vet J 2024; 305:106145. [PMID: 38788999 DOI: 10.1016/j.tvjl.2024.106145] [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: 09/18/2023] [Revised: 04/22/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Cancer is a major cause of death in humans and animals worldwide. While cancer survival rates have increased over recent decades, further research to identify risk factors for the onset and progression of disease, and safe and highly efficacious treatments, is needed. Spontaneous tumours in pets represent an excellent model for neoplastic disease in humans. In this regard, dogs are an interesting species, as the divergence between the dog and human genome is low, humans and dogs have important similarities in the development and functioning of the immune system, and both species often share the same physical environment. There is also a higher homology between the canine and human microbiome than murine model. This review aims to describe and organize recently published information on canine microbiome assemblages and their relationship with the onset and progression of colorectal cancer, breast cancer and lymphoma, and to compare this with human disease. In both species, dysbiosis can induce variations in the gut microbiota that strongly influence shifts in status between health and disease. This can produce an inflammatory state, potentially leading to neoplasia, especially in the intestine, thus supporting canine studies in comparative oncology. Intestinal dysbiosis can also alter the efficacy and side effects of cancer treatments. Fewer published studies are available on changes in the relevant microbiomes in canine lymphoma and mammary cancer, and further research in this area could improve our understanding of the role of microbiota in the development of these cancers.
Collapse
Affiliation(s)
- Dell'Anno Filippo
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Genova 16129, Italy; Department of Public Health, Experimental and Forensic Medicine, Section of Biostatistics and Clinical Epidemiology, University of Pavia, Pavia, Italy
| | - Lisa Guardone
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Genova 16129, Italy
| | - Valeria Listorti
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Genova 16129, Italy
| | - Razzuoli Elisabetta
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Genova 16129, Italy.
| |
Collapse
|
21
|
Alaba TE, Holman JM, Ishaq SL, Li Y. Current Knowledge on the Preparation and Benefits of Cruciferous Vegetables as Relates to In Vitro, In Vivo, and Clinical Models of Inflammatory Bowel Disease. Curr Dev Nutr 2024; 8:102160. [PMID: 38779039 PMCID: PMC11108850 DOI: 10.1016/j.cdnut.2024.102160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/01/2024] [Accepted: 04/14/2024] [Indexed: 05/25/2024] Open
Abstract
Inflammatory bowel disease is a chronic condition with a significant economic and social burden. The disease is complex and challenging to treat because it involves several pathologies, such as inflammation, oxidative stress, dysbiosis, and intestinal damage. The search for an effective treatment has identified cruciferous vegetables and their phytochemicals as potential management options for inflammatory bowel disease because they contain prebiotics, probiotics, and anti-inflammatory and antioxidant metabolites essential for a healthy gut. This critical narrative style review provides a robust insight into the pharmacological effects and benefits of crucifers and their documented bioactive compounds in in vitro and in vivo models, as well as clinical inflammatory bowel disease. The review highlights the significant impact of crucifer preparation and the presence of glucosinolates, isothiocyanates, flavonoids, and polyphenolic compounds, which are essential for the anti-inflammatory and antioxidative benefits of cruciferous vegetables, as well as their ability to promote the healthy microbial community and maintain the intestinal barrier. This review may serve as a viable nutritional guide for future research on methods and features essential to developing experiments, preventions, and treatments for inflammatory bowel disease. There is limited clinical information and future research may utilize current innovative tools, such as metabolomics, for adequate knowledge and effective translation into clinical therapy.
Collapse
Affiliation(s)
- Tolu E Alaba
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
| | - Johanna M Holman
- School of Food and Agriculture, University of Maine, Orono, ME, United States
| | - Suzanne L Ishaq
- School of Food and Agriculture, University of Maine, Orono, ME, United States
| | - Yanyan Li
- School of Food and Agriculture, University of Maine, Orono, ME, United States
- School of Pharmacy and Pharmaceutical Sciences, SUNY Binghamton University, Johnson City, NY, United States
| |
Collapse
|
22
|
Ding L, Liu J, Yang Y, Cui Z, Du G. Chronically socially isolated mice exhibit depressive-like behavior regulated by the gut microbiota. Heliyon 2024; 10:e29791. [PMID: 38681644 PMCID: PMC11046198 DOI: 10.1016/j.heliyon.2024.e29791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024] Open
Abstract
Objectives Chronic loneliness is a widespread issue, and the gut-brain axis is known to be crucial in facilitating communication between the gut and brain. However, the precise mechanism by which chronic loneliness affects the gut-brain axis remains uncertain. Methods Fourteen 55-week-old Balb/c mice were used in the experiment, with seven mice being randomly assigned to the chronic social isolation (CSI) group. The CSI group mice underwent 12 weeks of isolation to simulate the psychiatric state of a population in prolonged social isolation. The mental state of the CSI mice was assessed through animal behavior analysis, while plasma cytokines were measured using ELISA. Additionally, the composition of the gut microbiota was analyzed using 16S rRNA sequencing, and the metabolite composition of the intestinal contents was examined using nontargeted metabolomics. The Student-T test was used to determine significant mean differences. Results Mice that were exposed to the CSI exhibited increased immobility time lengths in forced swimming and hanging tail experiments, and decreased movement lengths and number of times traversing the intermediate region, compared to control mice. Additionally, CSI decreased the abundance of the probiotics Ruminococcaceae, Akkermansiaceae, and Christensenellaceae. Additionally, CSI reduced the production of the metabolites oleamide and tryptophan. Furthermore, IL-1β, IL-4, and IL-6 were significantly increased, while TNF-α was significantly decreased. Conclusion CSI induces a dysbiotic gut microbiota and the production of neurorelated metabolites, which in turn increase inflammatory responses and result in depressive behaviors in CSI mice. Therefore, these findings suggest that the gut microbiota may serve as a target for the treatment of long-term social isolation-induced mental disorders.
Collapse
Affiliation(s)
- Linwei Ding
- Biotechnology Laboratory, Hainan Medical University, Haikou, China
| | - Jiaqi Liu
- Biotechnology Laboratory, Hainan Medical University, Haikou, China
- Biotechnology Major, Hainan Medical University, Haikou, China
| | - Yunjia Yang
- Biotechnology Laboratory, Hainan Medical University, Haikou, China
- School of Public Health, Hainan Medical University, Haikou, China
| | - Zeying Cui
- Biotechnology Laboratory, Hainan Medical University, Haikou, China
| | - Guankui Du
- Biotechnology Laboratory, Hainan Medical University, Haikou, China
- Biotechnology Major, Hainan Medical University, Haikou, China
- Department of Biochemistry and Molecular Biology, Hainan Medical University, Haikou, China
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
23
|
Garrett S, Zhang Y, Xia Y, Sun J. Intestinal Epithelial Axin1 Deficiency Protects Against Colitis via Altered Gut Microbiota. ENGINEERING (BEIJING, CHINA) 2024; 35:241-256. [PMID: 38911180 PMCID: PMC11192507 DOI: 10.1016/j.eng.2023.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Intestinal homeostasis is maintained by specialized host cells and the gut microbiota. Wnt/β-catenin signaling is essential for gastrointestinal development and homeostasis, and its dysregulation has been implicated in inflammation and colorectal cancer. Axin1 negatively regulates activated Wnt/β-catenin signaling, but little is known regarding its role in regulating host-microbial interactions in health and disease. Here, we aim to demonstrate that intestinal Axin1 determines gut homeostasis and host response to inflammation. Axin1 expression was analyzed in human inflammatory bowel disease datasets. To explore the effects and mechanism of intestinal Axin1 in regulating intestinal homeostasis and colitis, we generated new mouse models with Axin1 conditional knockout in intestinal epithelial cell (IEC; Axin1 ΔIEC) and Paneth cell (PC; Axin1 ΔPC) to compare with control (Axin1 LoxP; LoxP: locus of X-over, P1) mice. We found increased Axin1 expression in the colonic epithelium of human inflammatory bowel disease (IBD). Axin1 ΔIEC mice exhibited altered goblet cell spatial distribution, PC morphology, reduced lysozyme expression, and enriched Akkermansia muciniphila (A. muciniphila). The absence of intestinal epithelial and PC Axin1 decreased susceptibility to dextran sulfate sodium (DSS)-induced colitis in vivo. Axin1 ΔIEC and Axin1 ΔPC mice became more susceptible to DSS-colitis after cohousing with control mice. Treatment with A. muciniphila reduced DSS-colitis severity. Antibiotic treatment did not change the IEC proliferation in the Axin1 Loxp mice. However, the intestinal proliferative cells in Axin1 ΔIEC mice with antibiotic treatment were reduced compared with those in Axin1 ΔIEC mice without treatment. These data suggest non-colitogenic effects driven by the gut microbiome. In conclusion, we found that the loss of intestinal Axin1 protects against colitis, likely driven by epithelial Axin1 and Axin1-associated A. muciniphila. Our study demonstrates a novel role of Axin1 in mediating intestinal homeostasis and the microbiota. Further mechanistic studies using specific Axin1 mutations elucidating how Axin1 modulates the microbiome and host inflammatory response will provide new therapeutic strategies for human IBD.
Collapse
Affiliation(s)
- Shari Garrett
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Yongguo Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Yinglin Xia
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Cancer Center, University of Illinois Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
24
|
Yang F, Lan Z, Chen H, He R. Causal associations between human gut microbiota and hemorrhoidal disease: A two-sample Mendelian randomization study. Medicine (Baltimore) 2024; 103:e37599. [PMID: 38552035 PMCID: PMC10977532 DOI: 10.1097/md.0000000000037599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/22/2024] [Indexed: 04/02/2024] Open
Abstract
Hemorrhoidal disease (HEM) is a common condition affecting a significant proportion of the population. However, the causal relationship between the gut microbiota and hemorrhoids remains unclear. In this study, we employed a Mendelian randomization (MR) approach to investigate the potential associations between them. In this study, the exposure factor was determined by selecting summary statistics data from a large-scale gut microbiome whole-genome association study conducted by the MiBioGen Consortium, which involved a sample size of 18,340 individuals. The disease outcome data consisted of 218,920 cases of HEM and 725,213 controls of European ancestry obtained from the European Bioinformatics Institute dataset. Two-sample MR analyses were performed to assess the causalities between gut microbiota and hemorrhoids using various methods, including inverse-variance weighting, MR-Egger regression, MR Pleiotropy Residual Sum and Outlier (MR-PRESSO), simple mode, and weighted median. Reverse MR analyses were performed to examine reverse causal association. Our findings suggest phylum Cyanobacteria (OR = 0.947, 95% CI: 0.915-0.980, P = 2.10 × 10 - 3), genus Phascolarctobacterium (OR = 0.960, 95% CI: 0.924-0.997, P = .034) and family FamilyXI (OR = 0.974, 95% CI: 0.952-0.997, P = .027) have potentially protective causal effects on the risk of HEM, while genus Ruminococcaceae_UCG_002 (OR = 1.036, 95% CI: 1.001-1.071, P = .042), family Peptostreptococcaceae (OR = 1.042, 95% CI: 1.004-1.082, P = .029), genus Oscillospira (OR = 1.048, 95% CI: 1.005-1.091, P = .026), family Alcaligenaceae (OR = 1.048, 95% CI: 1.005-1.091, P = .036) and order Burkholderiales (OR = 1.074, 95% CI: 1.020-1.130, P = 6.50 × 10-3) have opposite effect. However, there was a reverse causal relationship between HEM and genus Oscillospira (OR = 1.140, 95% CI: 1.002-1.295, P = .046) This is the first MR study to explore the causalities between specific gut microbiota taxa and hemorrhoidal disease, which may offer valuable insights for future clinical interventions for hemorrhoidal disease.
Collapse
Affiliation(s)
- Fang Yang
- Anorectal Department of Traditional Chinese Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhihua Lan
- Department of Pathology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Huabing Chen
- Anorectal Department of Traditional Chinese Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Rongfang He
- Department of Pathology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
25
|
Jang KK, Ercelen D, Cen Feng JYC, Gurunathan S, Zhou C, Korman A, Newell L, Hudesman D, Jones DR, Loke P, Axelrad JE, Cadwell K. Tofacitinib uptake by patient-derived intestinal organoids predicts individual clinical responsiveness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.02.583137. [PMID: 38712048 PMCID: PMC11071318 DOI: 10.1101/2024.03.02.583137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Background & Aims Despite increasing therapeutic options in the treatment of ulcerative colitis (UC), achieving disease remission remains a major clinical challenge. Nonresponse to therapy is common and clinicians have little guidance in selecting the optimal therapy for an individual patient. This study examined whether patient-derived materials could predict individual clinical responsiveness to the Janus kinase (JAK) inhibitor, tofacitinib, prior to treatment initiation. Method In 48 patients with UC initiating tofacitinib, we longitudinally collected clinical covariates, stool, and colonic biopsies to analyze the microbiota, transcriptome, and exome variations associated with clinical responsiveness at week 24. We established patient-derived organoids (n = 23) to determine how their viability upon stimulation with proinflammatory cytokines in the presence of tofacitinib related to drug responsiveness in patients. We performed additional biochemical analyses of organoids and primary tissues to identify the mechanism underlying differential tofacitinib sensitivity. Results The composition of the gut microbiota, rectal transcriptome, inflammatory biomarkers, and exome variations were indistinguishable among UC patients prior to tofacitinib treatment. However, a subset of patient-derived organoids displayed reduced sensitivity to tofacitinib as determined by the ability of the drug to inhibit STAT1 phosphorylation and loss of viability upon cytokine stimulation. Remarkably, sensitivity of organoids to tofacitinib predicted individual clinical patient responsiveness. Reduced responsiveness to tofacitinib was associated with decreased levels of the cationic transporter MATE1, which mediates tofacitinib uptake. Conclusions Patient-derived intestinal organoids predict and identify mechanisms of individual tofacitinib responsiveness in UC. Specifically, MATE1 expression predicted clinical response to tofacitinib.
Collapse
|
26
|
Ye W, Fan J, Wu W, Chen Z, Huang Q, Qian L. Effects of fecal microbiota transplantation on metabolic health of DBA mice. Front Microbiol 2024; 15:1352555. [PMID: 38444807 PMCID: PMC10912182 DOI: 10.3389/fmicb.2024.1352555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/31/2024] [Indexed: 03/07/2024] Open
Abstract
Introduction Numerous studies have demonstrated that C57BL/6 mice exhibit superior growth rates and overall growth performance compared to DBA mice. To investigate whether this discrepancy in growth performance is linked to the composition of gut microorganisms, we conducted fecal microbiome transplantation (FMT) experiments. Methods Specifically, we transplanted fecal fluids from adult C57BL/6 mice, high-fat C57BL/6 mice, and Wistar rats into weaned DBA mice (0.2mL/d), and subsequently analyzed their gut contents and gene expression through 16S rRNA sequencing and transcriptome sequencing. During the test period, C57BL/6 mice and Wistar rats were provided with a normal diet, and high-fat C57BL/6 mice were provided with a high-fat diet. Results The results of our study revealed that mice receiving FMT from all three donor groups exhibited significantly higher daily weight gain and serum triglyceride (TG) levels compared to mice of CK group. 16S rRNA sequensing unveiled substantial differences in the abundance and function of the gut microbiota between the FMT groups and the CK group. Transcriptome analysis revealed a total of 988 differential genes, consisting of 759 up-regulated genes and 187 down-regulated genes, between the three experimental groups and the CK group. Functional Gene Ontology (GO) annotation suggested that these genes were primarily linked to lipid metabolism, coagulation, and immunity. Pearson correlation analysis was performed on the differential genes and clusters, and it revealed significant correlations, mainly related to processes such as fatty acid metabolism, fat digestion and absorption, and cholesterol metabolism. Discussion In summary, FMT from dominant strains improved the growth performance of DBA mice, including body weight gain, institutional growth, and immune performance. This change may be due to the increase of probiotic content in the intestinal tract by FMT and subsequent alteration of intestinal gene expression. However, the effects of cross-species fecal transplantation on the intestinal flora and gene expression of recipient mice were not significant.
Collapse
Affiliation(s)
- Wenxin Ye
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jinghui Fan
- Hangzhou Academy of Agricultural Sciences, Hangzhou, China
| | - Wenzi Wu
- Hainan Institute of Zhejiang University, Sanya, China
| | - Zhuo Chen
- Hainan Institute of Zhejiang University, Sanya, China
| | - Qixin Huang
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Lichun Qian
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
27
|
Chen H, Qian Y, Jiang C, Tang L, Yu J, Zhang L, Dai Y, Jiang G. Butyrate ameliorated ferroptosis in ulcerative colitis through modulating Nrf2/GPX4 signal pathway and improving intestinal barrier. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166984. [PMID: 38061600 DOI: 10.1016/j.bbadis.2023.166984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023]
Abstract
Oxidative stress and intestinal inflammation are main pathological features of ulcerative colitis (UC). Ferroptosis, characterized by iron accumulation and lipid peroxidation, is closely related to the pathologic process of UC. 16S rRNA sequencing for intestinal microbiota analysis and gas chromatography-mass spectrometry (GC-MS) for short-chain fatty acid (SCFA) contents clearly demonstrated lower amounts of butyrate-producing bacteria and butyrate in colitis mice. However, the precise mechanisms of sodium butyrate (NaB) in treating UC remain largely unclear. We found that ferroptosis occurred in colitis models, as evidenced by the inflammatory response, intracellular iron level, mitochondria ultrastructural observations and associated protein expression. NaB inhibited ferroptosis in colitis, significantly rescued weight loss and colon shortening in mice and reduced inflammatory lesions and mitochondrial damage. Furthermore, NaB improved intestinal barrier integrity and markedly suppressed the expression of pro-ferroptosis proteins. Conversely, the protein expression of anti-ferroptosis markers including nuclear factor erythroid-related Factor 2 (Nrf2) and glutathione peroxidase 4 (GPX4), was significantly upregulated with NaB treatment. Moreover, the knockdown of Nrf2 reversed the anti-colitis effect of NaB. Taken together, NaB exhibited a protective effect by ameliorating ferroptosis in experimental colitis through Nrf2/GPX4 signaling and improving intestinal barrier integrity, which provides a novel mechanism for NaB prevention of UC.
Collapse
Affiliation(s)
- Hangping Chen
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou 311200, Zhejiang, China
| | - Yifan Qian
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang, China
| | - Chensheng Jiang
- Department of Gastroenterology, The Fourth Affiliated Hospital, College of Medicine, Zhejiang University, Yiwu 322099, Zhejiang, China
| | - Leilei Tang
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou 311200, Zhejiang, China
| | - Jiawen Yu
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou 311200, Zhejiang, China
| | - Lingdi Zhang
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou 311200, Zhejiang, China
| | - Yiyang Dai
- Department of Gastroenterology, The Fourth Affiliated Hospital, College of Medicine, Zhejiang University, Yiwu 322099, Zhejiang, China.
| | - Guojun Jiang
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou 311200, Zhejiang, China.
| |
Collapse
|
28
|
Paiano L, Mastronardi M, Campisciano G, Rosso N, Casagranda B, Comar M, de Manzini N, Palmisano S. Liver Bacterial Colonization in Patients with Obesity and Gut Dysbiosis. Obes Surg 2024; 34:402-408. [PMID: 38102371 DOI: 10.1007/s11695-023-06989-2] [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: 10/01/2023] [Revised: 11/27/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
PURPOSE Recently, the link between gut microbiota, liver inflammation, and obesity has become an interesting focus of research. The aim of this study is to show the possible relation between gut microbiota dysbiosis in patients with obesity and the presence of bacterial genomes in their liver biopsies. MATERIALS AND METHODS A prospective study on patients undergoing bariatric surgery was carried out. Anthropometric and metabolic data, comorbidities, stool samples, and hepatic biopsies were collected and analyzed at the time of surgery. The V3-16S rRNA region was sequenced using the Ion Torrent new-generation sequencing platform. RESULTS In each of the 23 patients enrolled, the bacterial population was analyzed both in the stools and liver. In eight patients (34.7%), Prevotella (62.5%), Bacteroides (50%), Streptococcus (12.5%), and Dalister (12.5%) were found in both samples, simultaneously; in 15 cases, the liver was free from colonization. The statistically significant difference between groups was a Roseburia intestinalis reduction in fecal samples of patients with liver biopsies colonized by bacteria (1% vs 3%; p = 0.0339). CONCLUSION To the best of our knowledge, this is the first study reporting the presence of bacterial genome in a liver biopsy on bariatric patients, instead of the microbe-associated molecular patterns. Notably, in literature, the presence of Roseburia intestinalis in stool samples has been shown to prevent intestinal inflammation playing its role in the gut barrier integrity. In our population, the Roseburia reduction was associated with the presence of bacterial genome in the liver, probably related to a greater permeability of the gut and vascular barriers.
Collapse
Affiliation(s)
- Lucia Paiano
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy
| | - Manuela Mastronardi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy.
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy.
| | - Giuseppina Campisciano
- Institute for Maternal and Child Health IRCCS, Burlo Garofolo, Via dell' Istria 65/1, 34149, Trieste, Italy
| | - Natalia Rosso
- Fondazione Italiana Fegato, Centro Studi Fegato, Area Science Park Basovizza Bldg.Q SS14 Km, 163.5, 34149, Trieste, Italy
| | - Biagio Casagranda
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy
| | - Manola Comar
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy
- Institute for Maternal and Child Health IRCCS, Burlo Garofolo, Via dell' Istria 65/1, 34149, Trieste, Italy
| | - Nicolò de Manzini
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy
| | - Silvia Palmisano
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy
| |
Collapse
|
29
|
Liu C, Qi X, Li D, Zhao L, Li Q, Mao K, Shen G, Ma Y, Wang R. Limosilactobacillus fermentum HF06-derived paraprobiotic and postbiotic alleviate intestinal barrier damage and gut microbiota disruption in mice with ulcerative colitis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:1702-1712. [PMID: 37851615 DOI: 10.1002/jsfa.13057] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/16/2023] [Accepted: 10/19/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND Paraprobiotics and postbiotics have shown potential in the treatment of ulcerative colitis (UC). However, their in vivo application is still in its infancy and their mechanisms of action are not well understood. RESULTS Here, we investigated the mitigation effects of Limosilactobacillus fermentum HF06-derived paraprobiotic (6-PA) and postbiotic (6-PS) on dextran sulfate sodium induced UC and the potential mechanisms. Results indicated that the administration of 6-PA and 6-PS resulted in the inhibition of weight loss and colon shortening in mice with UC. Furthermore, they led to a significant reduction in both fecal moisture content and the levels of proinflammatory cytokines and oxidative stress in the intestine of the mice. 6-PA and 6-PS treatment strengthened the intestinal mucosal barrier by dramatically upregulating the levels of zonula occludens-1 and occludin proteins. In addition, 6-PA and 6-PS restored intestinal dysbiosis by regulating abundances of certain bacteria, such as Bifidobacterium, Faecalibaculum, Muribaculaceae, Corynebacterium, Escherichia-Shigella and Clostridium_sensu_stricto_1, and regulated the level of short-chain fatty acids. CONCLUSION These findings illustrated for the first time that L. fermentum HF06-derived paraprobiotic and postbiotic enhanced the intestinal barrier function, and restored gut microbiota alterations. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Chunhong Liu
- School of Chemical Engineering and Chemistry, Harbin Institute of Technology, Harbin, China
| | - Xiaofen Qi
- School of Chemical Engineering and Chemistry, Harbin Institute of Technology, Harbin, China
| | - Dan Li
- School of Chemical Engineering and Chemistry, Harbin Institute of Technology, Harbin, China
| | - Le Zhao
- School of Chemical Engineering and Chemistry, Harbin Institute of Technology, Harbin, China
| | - Qiming Li
- Dairy Nutrition and Function, Key Laboratory of Sichuan Province, New Hope Dairy Company Limited, Chengdu, China
| | - Kaidong Mao
- Jiangsu HOWYOU Biotechnology Co. Ltd, Qidong, China
| | - Guiqi Shen
- Jiangsu HOWYOU Biotechnology Co. Ltd, Qidong, China
| | - Ying Ma
- School of Chemical Engineering and Chemistry, Harbin Institute of Technology, Harbin, China
| | - Rongchun Wang
- School of Chemical Engineering and Chemistry, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
30
|
Troci A, Philippen S, Rausch P, Rave J, Weyland G, Niemann K, Jessen K, Schmill LP, Aludin S, Franke A, Berg D, Bang C, Bartsch T. Disease- and stage-specific alterations of the oral and fecal microbiota in Alzheimer's disease. PNAS NEXUS 2024; 3:pgad427. [PMID: 38205031 PMCID: PMC10776369 DOI: 10.1093/pnasnexus/pgad427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 11/30/2023] [Indexed: 01/12/2024]
Abstract
Microbial communities in the intestinal tract are suggested to impact the ethiopathogenesis of Alzheimer's disease (AD). The human microbiome might modulate neuroinflammatory processes and contribute to neurodegeneration in AD. However, the microbial compositions in patients with AD at different stages of the disease are still not fully characterized. We used 16S rRNA analyses to investigate the oral and fecal microbiota in patients with AD and mild cognitive impairment (MCI; n = 84), at-risk individuals (APOE4 carriers; n = 17), and healthy controls (n = 50) and investigated the relationship of microbial communities and disease-specific markers via multivariate- and network-based approaches. We found a slightly decreased diversity in the fecal microbiota of patients with AD (average Chao1 diversity for AD = 212 [SD = 66]; for controls = 215 [SD = 55]) and identified differences in bacterial abundances including Bacteroidetes, Ruminococcus, Sutterella, and Porphyromonadaceae. The diversity in the oral microbiota was increased in patients with AD and at-risk individuals (average Chao1 diversity for AD = 174 [SD = 60], for at-risk group = 195 [SD = 49]). Gram-negative proinflammatory bacteria including Haemophilus, Neisseria, Actinobacillus, and Porphyromonas were dominant oral bacteria in patients with AD and MCI and the abundance correlated with the cerebrospinal fluid biomarker. Taken together, we observed a strong shift in the fecal and the oral communities of patients with AD already prominent in prodromal and, in case of the oral microbiota, in at-risk stages. This indicates stage-dependent alterations in oral and fecal microbiota in AD which may contribute to the pathogenesis via a facilitated intestinal and systemic inflammation leading to neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Alba Troci
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Sarah Philippen
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Philipp Rausch
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Julius Rave
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Gina Weyland
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Katharina Niemann
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Katharina Jessen
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Lars-Patrick Schmill
- Department of Radiology and Neuroradiology, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Schekeb Aludin
- Department of Radiology and Neuroradiology, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Daniela Berg
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Thorsten Bartsch
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| |
Collapse
|
31
|
Kiely LJ, Busca K, Lane JA, van Sinderen D, Hickey RM. Molecular strategies for the utilisation of human milk oligosaccharides by infant gut-associated bacteria. FEMS Microbiol Rev 2023; 47:fuad056. [PMID: 37793834 PMCID: PMC10629584 DOI: 10.1093/femsre/fuad056] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 09/14/2023] [Accepted: 10/03/2023] [Indexed: 10/06/2023] Open
Abstract
A number of bacterial species are found in high abundance in the faeces of healthy breast-fed infants, an occurrence that is understood to be, at least in part, due to the ability of these bacteria to metabolize human milk oligosaccharides (HMOs). HMOs are the third most abundant component of human milk after lactose and lipids, and represent complex sugars which possess unique structural diversity and are resistant to infant gastrointestinal digestion. Thus, these sugars reach the infant distal intestine intact, thereby serving as a fermentable substrate for specific intestinal microbes, including Firmicutes, Proteobacteria, and especially infant-associated Bifidobacterium spp. which help to shape the infant gut microbiome. Bacteria utilising HMOs are equipped with genes associated with their degradation and a number of carbohydrate-active enzymes known as glycoside hydrolase enzymes have been identified in the infant gut, which supports this hypothesis. The resulting degraded HMOs can also be used as growth substrates for other infant gut bacteria present in a microbe-microbe interaction known as 'cross-feeding'. This review describes the current knowledge on HMO metabolism by particular infant gut-associated bacteria, many of which are currently used as commercial probiotics, including the distinct strategies employed by individual species for HMO utilisation.
Collapse
Affiliation(s)
- Leonie Jane Kiely
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61C996, Ireland
- Health and Happiness Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork P61K202, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - Kizkitza Busca
- Health and Happiness Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork P61K202, Ireland
| | - Jonathan A Lane
- Health and Happiness Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork P61K202, Ireland
| | - Douwe van Sinderen
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - Rita M Hickey
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61C996, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
| |
Collapse
|
32
|
Di Ciaula A, Bonfrate L, Khalil M, Garruti G, Portincasa P. Contribution of the microbiome for better phenotyping of people living with obesity. Rev Endocr Metab Disord 2023; 24:839-870. [PMID: 37119391 PMCID: PMC10148591 DOI: 10.1007/s11154-023-09798-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 05/01/2023]
Abstract
Obesity has reached epidemic proportion worldwide and in all ages. Available evidence points to a multifactorial pathogenesis involving gene predisposition and environmental factors. Gut microbiota plays a critical role as a major interface between external factors, i.e., diet, lifestyle, toxic chemicals, and internal mechanisms regulating energy and metabolic homeostasis, fat production and storage. A shift in microbiota composition is linked with overweight and obesity, with pathogenic mechanisms involving bacterial products and metabolites (mainly endocannabinoid-related mediators, short-chain fatty acids, bile acids, catabolites of tryptophan, lipopolysaccharides) and subsequent alterations in gut barrier, altered metabolic homeostasis, insulin resistance and chronic, low-grade inflammation. Although animal studies point to the links between an "obesogenic" microbiota and the development of different obesity phenotypes, the translational value of these results in humans is still limited by the heterogeneity among studies, the high variation of gut microbiota over time and the lack of robust longitudinal studies adequately considering inter-individual confounders. Nevertheless, available evidence underscores the existence of several genera predisposing to obesity or, conversely, to lean and metabolically health phenotype (e.g., Akkermansia muciniphila, species from genera Faecalibacterium, Alistipes, Roseburia). Further longitudinal studies using metagenomics, transcriptomics, proteomics, and metabolomics with exact characterization of confounders are needed in this field. Results must confirm that distinct genera and specific microbial-derived metabolites represent effective and precision interventions against overweight and obesity in the long-term.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Gabriella Garruti
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| |
Collapse
|
33
|
Liu L, Zhang S, Zhi F, Song Y, Li B, Gao P, Zhang Y, Ma K, Xu J, Jiang B, Chu Y, Li Y, Qin J. RNA helicase DExD/H-box 5 modulates intestinal microbiota in mice. Microb Pathog 2023; 182:106265. [PMID: 37482112 DOI: 10.1016/j.micpath.2023.106265] [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: 06/19/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 07/25/2023]
Abstract
The RNA helicase DExD/H-box (DDX) family of proteins plays a central role in host cellular RNA metabolism, including mRNA regulation, microRNA biogenesis, and ribosomal processing. DDX5, also known as p68, promotes viral replication and tumorigenesis. However, there have been no studies on the regulation of the intestinal microbiota by DDX family proteins. We constructed DDX5 knockout mice (Ddx5+/-) using CRISPR/CAS9 technology. Subsequently, DDX5 knockout mice were analyzed for PCR products, mRNA levels, protein expression, immunohistochemistry, and histopathological lesions. Fecal (n = 12) and ileum (n = 12) samples were collected from the Ddx5+/- and wild-type (Ddx5+/+) mice. The diversity, richness, and structural separation of the intestinal microbiota of the Ddx5+/- and Ddx5+/+ mice were determined by 16S rRNA sequencing and analysis. Ddx5+/- mice were successfully established, and the ileum had normal morphology, a clear layer of tissue structures, and neatly arranged cupped cells. DDX5 knockout mice did not exhibit adverse effects on the ileal tissue. Microbial diversity and abundance were not significantly different, but the microbial structure of the intestinal microbiota was clustered separately between Ddx5+/+ and Ddx5+/- mice. Furthermore, we found that the relative abundance of Akkermansia and Clostridium_sensu_stricto_1 in the Ddx5+/- mice was significantly lower than in the Ddx5+/+ mice. These analyses indicated specific interactions between the intestinal microbiota and DDX5 protein. Our results indicate that DDX5 has a significant effect on the composition of the intestinal microbiota in mice, suggesting its potential as a promising novel target for the treatment of inflammation and tumorigenesis in the intestine.
Collapse
Affiliation(s)
- Liyuan Liu
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei Province, 071001, China; State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China
| | - Silan Zhang
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang Province, 830091, China
| | - Feijie Zhi
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China
| | - Yinjuan Song
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China
| | - Bin Li
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang Province, 830091, China
| | - Pengchen Gao
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China
| | - Ying Zhang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China
| | - Ke Ma
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China
| | - Jian Xu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang Province, 830091, China
| | - Bo Jiang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing, 100097, China
| | - Yuefeng Chu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang Province, 830091, China
| | - Yongqing Li
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing, 100097, China
| | - Jianhua Qin
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei Province, 071001, China.
| |
Collapse
|
34
|
Ahmed EA, Ahmed SM, Zakaria NH, Baddour NM, Header DA. Study of the gut microbiome in Egyptian patients with active ulcerative colitis. REVISTA DE GASTROENTEROLOGIA DE MEXICO (ENGLISH) 2023; 88:246-255. [PMID: 35906158 DOI: 10.1016/j.rgmxen.2022.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 10/13/2021] [Indexed: 11/24/2022]
Abstract
INTRODUCTION AND AIM Ulcerative colitis (UC) is characterized by chronic, uncontrolled inflammation of the intestinal mucosa. Gut microbiota dysbiosis was reported to be a factor in intestinal inflammation. The aim of the present study was to study changes in the gut microbiome in Egyptian patients with active UC. MATERIALS AND METHODS In this cross-sectional study, the gut bacterial microbiome of 21 UC patients and 20 control subjects was analyzed using the quantitative SYBR Green real-time PCR technique, targeting the 16S rRNA gene of selected bacterial phyla/genera and/or species. RESULTS UC patients showed marked dysbiosis evidenced by a significant decrease in the Firmicutes and F. prausnitzii anti-inflammatory bacteria. The Firmicutes/Bacteroidetes ratio was also lower in the UC cases (1.65), compared with the healthy controls (2.93). In addition, the UC cases showed a statistically significant decrease in Ruminococcus, compared with the control group. However, there were no statistically significant differences between UC patients and the controls, regarding A. muciniphila, Bifidobacterium, Lactobacillus, Bacteroides, and Prevotella. One UC case was positive for the pathogenic bacterium, Clostridioides difficile, with low relative abundance. CONCLUSION The current study showed differences in the gut microbiome of UC patients, compared with healthy controls. This may help in identifying the gut microbiome and specific bacterial changes that can be targeted for treatment of UC.
Collapse
Affiliation(s)
- E A Ahmed
- Departamento de Medicina Interna, Unidad de Gastroenterología, Facultad de Medicina, Universidad de Alejandría, Alejandría, Egypt
| | - S M Ahmed
- Departamento de Microbiología e Inmunología Médica, Facultad de Medicina, Universidad de Alejandría, Alejandría, Egypt
| | - N H Zakaria
- Departamento de Patología Clínica, Facultad de Medicina, Universidad de Alejandría, Alejandría, Egypt
| | - N M Baddour
- Departamento de Patología, Facultad de Medicina, Universidad de Alejandría, Alejandría, Egypt
| | - D A Header
- Departamento de Medicina Interna, Unidad de Gastroenterología, Facultad de Medicina, Universidad de Alejandría, Alejandría, Egypt.
| |
Collapse
|
35
|
Al-Bayati L, Fasaei BN, Merat S, Bahonar A, Ghoddusi A. Quantitative analysis of the three gut microbiota in UC and non-UC patients using real-time PCR. Microb Pathog 2023:106198. [PMID: 37295481 DOI: 10.1016/j.micpath.2023.106198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND and study aims: Gastrointestinal microbiota are closely related to the pathogenesis of ulcerative colitis (UC). This study aimed at quantification of F. prausnitzii, Provetella, and Peptostreptococcus in UC and non-UC patients using Real-Time PCR and a new set of primers were also validated for this purpose. MATERIALS AND METHODS In this study, the relative abundance of microbial populations between the UC and non-UC subjects were evaluated by quantitative real-time polymerase chain reaction (qRT-PCR). DNA extraction from biopsies and polymerase chain reaction (PCR) amplification of bacterial 16S rRNA gene-targeted species-specific primers was performed to detect the anaerobic bacterial species. The qRT-PCR was used to show the relative change in the bacterial populations of F. prausnitzii, Provetella, and Peptostreptococcus in the UC and non-UC subjects. RESULTS Our data for detection of the anaerobic intestinal flora showed Faecalibacterium prausnitzii, Provetella and Peptostreptococcus were the predominant microflora in the controls and showed significant differences (p = 0.002, 0.025 and 0.039, respectively). The qRT-PCR analyses of F. prausnitzii, Provetella and Peptostreptococcus were 8.69-, 9.38- and 5.77-higher, respectively, in the control group than in the UC group. CONCLUSION The results of this study showed decreased abundance of F. prausnitzii, Provetella and Peptostreptococcus in the intestine of UC patients in comparison to non-UC patients. Quantitative RT-PCR, as a progressive and sensitive method, could be useful for evaluation of bacterial populations in patients with inflammatory bowel diseases to attain appropriate therapeutic strategies.
Collapse
Affiliation(s)
- Luma Al-Bayati
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran; Department of Microbiology, Faculty of Medicine, University of Wassit, Iraq
| | - Bahar Nayeri Fasaei
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Shahin Merat
- Digestive Disease Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Liver and Pancreatobiliary Diseases Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Bahonar
- Department of Food Hygiene, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Arefeh Ghoddusi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
36
|
Wade H, Pan K, Duan Q, Kaluzny S, Pandey E, Fatumoju L, Saraswathi V, Wu R, Harris EN, Su Q. Akkermansia muciniphila and its membrane protein ameliorates intestinal inflammatory stress and promotes epithelial wound healing via CREBH and miR-143/145. J Biomed Sci 2023; 30:38. [PMID: 37287024 PMCID: PMC10249216 DOI: 10.1186/s12929-023-00935-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 05/30/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND The intestinal epithelial barrier is the interface for interaction between gut microbiota and host metabolic systems. Akkermansia muciniphila (A. muciniphila) is a key player in the colonic microbiota that resides in the mucus layer, whose abundance is selectively decreased in the faecal microbiota of inflammatory bowel disease (IBD) patients. This study aims to investigate the regulatory mechanism among A. muciniphila, a transcription factor cAMP-responsive element-binding protein H (CREBH), and microRNA-143/145 (miR-143/145) in intestinal inflammatory stress, gut barrier integrity and epithelial regeneration. METHODS A novel mouse model with increased colonization of A muciniphila in the intestine of CREBH knockout mice, an epithelial wound healing assay and several molecular biological techniques were applied in this study. Results were analysed using a homoscedastic 2-tailed t-test. RESULTS Increased colonization of A. muciniphila in mouse gut enhanced expression of intestinal CREBH, which was associated with the mitigation of intestinal endoplasmic reticulum (ER) stress, gut barrier leakage and blood endotoxemia induced by dextran sulfate sodium (DSS). Genetic depletion of CREBH (CREBH-KO) significantly inhibited the expression of tight junction proteins that are associated with gut barrier integrity, including Claudin5 and Claudin8, but upregulated Claudin2, a tight junction protein that enhances gut permeability, resulting in intestinal hyperpermeability and inflammation. Upregulation of CREBH by A. muciniphila further coupled with miR-143/145 promoted intestinal epithelial cell (IEC) regeneration and wound repair via insulin-like growth factor (IGF) and IGFBP5 signalling. Moreover, the gene expressing an outer membrane protein of A. muciniphila, Amuc_1100, was cloned into a mammalian cell-expression vector and successfully expressed in porcine and human IECs. Expression of Amuc_1100 in IECs could recapitulate the health beneficial effect of A. muciniphila on the gut by activating CREBH, inhibiting ER stress and enhancing the expression of genes involved in gut barrier integrity and IEC's regeneration. CONCLUSIONS This study uncovers a novel mechanism that links A. muciniphila and its membrane protein with host CREBH, IGF signalling and miRNAs in mitigating intestinal inflammatory stress-gut barrier permeability and promoting intestinal wound healing. This novel finding may lend support to the development of therapeutic approaches for IBD by manipulating the interaction between host genes, gut bacteria and its bioactive components.
Collapse
Affiliation(s)
- Henry Wade
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5DL, UK
| | - Kaichao Pan
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, USA
| | - Qihua Duan
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5DL, UK
| | - Szczepan Kaluzny
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5DL, UK
| | - Ekta Pandey
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Linda Fatumoju
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | | | - Rongxue Wu
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, USA
| | - Edward N Harris
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Qiaozhu Su
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5DL, UK.
| |
Collapse
|
37
|
Jeong JJ, Ganesan R, Jin YJ, Park HJ, Min BH, Jeong MK, Yoon SJ, Choi MR, Choi J, Moon JH, Min U, Lim JH, Lee DY, Han SH, Ham YL, Kim BY, Suk KT. Multi-strain probiotics alleviate loperamide-induced constipation by adjusting the microbiome, serotonin, and short-chain fatty acids in rats. Front Microbiol 2023; 14:1174968. [PMID: 37333632 PMCID: PMC10272585 DOI: 10.3389/fmicb.2023.1174968] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Constipation is one of the most common gastrointestinal (GI) disorders worldwide. The use of probiotics to improve constipation is well known. In this study, the effect on loperamide-induced constipation by intragastric administration of probiotics Consti-Biome mixed with SynBalance® SmilinGut (Lactobacillus plantarum PBS067, Lactobacillus rhamnosus LRH020, Bifidobacterium animalis subsp. lactis BL050; Roelmi HPC), L. plantarum UALp-05 (Chr. Hansen), Lactobacillus acidophilus DDS-1 (Chr. Hansen), and Streptococcus thermophilus CKDB027 (Chong Kun Dang Bio) to rats was evaluated. To induce constipation, 5 mg/kg loperamide was intraperitoneally administered twice a day for 7 days to all groups except the normal control group. After inducing constipation, Dulcolax-S tablets and multi-strain probiotics Consti-Biome were orally administered once a day for 14 days. The probiotics were administered 0.5 mL at concentrations of 2 × 108 CFU/mL (G1), 2 × 109 CFU/mL (G2), and 2 × 1010 CFU/mL (G3). Compared to the loperamide administration group (LOP), the multi-strain probiotics not only significantly increased the number of fecal pellets but also improved the GI transit rate. The mRNA expression levels of serotonin- and mucin-related genes in the colons that were treated with the probiotics were also significantly increased compared to levels in the LOP group. In addition, an increase in serotonin was observed in the colon. The cecum metabolites showed a different pattern between the probiotics-treated groups and the LOP group, and an increase in short-chain fatty acids was observed in the probiotic-treated groups. The abundances of the phylum Verrucomicrobia, the family Erysipelotrichaceae and the genus Akkermansia were increased in fecal samples of the probiotic-treated groups. Therefore, the multi-strain probiotics used in this experiment were thought to help alleviate LOP-induced constipation by altering the levels of short-chain fatty acids, serotonin, and mucin through improvement in the intestinal microflora.
Collapse
Affiliation(s)
- Jin-Ju Jeong
- Institute for Liver and Digestive Disease, Hallym University, Chuncheon, Republic of Korea
| | - Raja Ganesan
- Institute for Liver and Digestive Disease, Hallym University, Chuncheon, Republic of Korea
| | - Yoo-Jeong Jin
- R&D Center, Chong Kun Dang Healthcare, Seoul, Republic of Korea
| | - Hee Jin Park
- Institute for Liver and Digestive Disease, Hallym University, Chuncheon, Republic of Korea
| | - Byeong Hyun Min
- Institute for Liver and Digestive Disease, Hallym University, Chuncheon, Republic of Korea
| | - Min Kyo Jeong
- Institute for Liver and Digestive Disease, Hallym University, Chuncheon, Republic of Korea
| | - Sang Jun Yoon
- Institute for Liver and Digestive Disease, Hallym University, Chuncheon, Republic of Korea
| | - Mi Ran Choi
- Institute for Liver and Digestive Disease, Hallym University, Chuncheon, Republic of Korea
| | - Jieun Choi
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute of Agricultural and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ji Hyun Moon
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute of Agricultural and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Uigi Min
- R&D Center, Chong Kun Dang Healthcare, Seoul, Republic of Korea
| | - Jong-Hyun Lim
- R&D Center, Chong Kun Dang Healthcare, Seoul, Republic of Korea
| | - Do Yup Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute of Agricultural and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang Hak Han
- Department of Pathology, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Young Lim Ham
- Department of Nursing, Daewon University College, Jecheon, Republic of Korea
| | - Byung-Yong Kim
- R&D Center, Chong Kun Dang Healthcare, Seoul, Republic of Korea
| | - Ki Tae Suk
- Institute for Liver and Digestive Disease, Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
38
|
Ikegami S, Nakamura M, Honda T, Yamamura T, Maeda K, Sawada T, Ishikawa E, Yamamoto K, Furune S, Ishikawa T, Furukawa K, Ohno E, Ishigami M, Kinoshita F, Kadota Y, Tochio T, Shimomura Y, Hirooka Y, Kawashima H. Efficacy of 1-kestose supplementation in patients with mild to moderate ulcerative colitis: A randomised, double-blind, placebo-controlled pilot study. Aliment Pharmacol Ther 2023; 57:1249-1257. [PMID: 36644995 DOI: 10.1111/apt.17387] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/21/2022] [Accepted: 12/29/2022] [Indexed: 01/17/2023]
Abstract
BACKGROUND Ulcerative colitis involves an excessive immune response to intestinal bacteria. Whether administering prebiotic 1-kestose is effective for active ulcerative colitis remains controversial. AIMS This randomised, double-blind, placebo-controlled pilot trial investigated the efficacy of 1-kestose against active ulcerative colitis. METHODS Forty patients with mild to moderate active ulcerative colitis were randomly treated with 1-kestose (N = 20) or placebo (maltose, N = 20) orally for 8 weeks in addition to the standard treatment. The Lichtiger clinical activity index and Ulcerative Colitis Endoscopic Index of Severity were determined. Faecal samples were analysed to evaluate the gut microbiome and metabolites. RESULTS The clinical activity index at week 8 was significantly lower in the 1-kestose group than in the placebo group (3.8 ± 2.7 vs. 5.6 ± 2.1, p = 0.026). Clinical remission and response rates were higher in the 1-kestose group than in the placebo group (remission: 55% vs. 20%, p = 0.048; response: 60% vs. 25%, p = 0.054). The Ulcerative Colitis Endoscopic Index of Severity at week 8 was not significantly different (2.8 ± 1.6 vs. 3.5 ± 1.6, p = 0.145). Faecal analysis showed significantly reduced alpha-diversity in the 1-kestose group, with a decreased relative abundance of several bacteria, including Ruminococcus gnavus group. The short-chain fatty acid levels were not significantly different between the groups. The incidence of adverse events was comparable between the groups. DISCUSSION Oral 1-kestose is well tolerated and provides clinical improvement for patients with mild to moderate ulcerative colitis through modulation of the gut microbiome.
Collapse
Affiliation(s)
- Shuji Ikegami
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masanao Nakamura
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Honda
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeshi Yamamura
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keiko Maeda
- Department of Endoscopy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tsunaki Sawada
- Department of Endoscopy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Eri Ishikawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenta Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Furune
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takuya Ishikawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhiro Furukawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Eizaburo Ohno
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masatoshi Ishigami
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumie Kinoshita
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | | | | | - Yoshiharu Shimomura
- Department of Food and Nutritional Sciences, College of Bioscience and Biotechnology, Chubu University, Kasugai, Japan
| | - Yoshiki Hirooka
- Department of Liver, Biliary Tract, and Pancreas Diseases, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
39
|
Segers A, de Vos WM. Mode of action of Akkermansia muciniphila in the intestinal dialogue: role of extracellular proteins, metabolites and cell envelope components. MICROBIOME RESEARCH REPORTS 2023; 2:6. [PMID: 38045608 PMCID: PMC10688800 DOI: 10.20517/mrr.2023.05] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/01/2023] [Accepted: 03/07/2023] [Indexed: 12/05/2023]
Abstract
Akkermansia muciniphila is a promising next-generation beneficial microbe due to its natural presence in the mucus layer of the gut, its symbiotic ability to degrade mucus, and its capacity to improve the intestinal barrier function. A. muciniphila is able to counteract weight gain and immuno-metabolic disturbances in several animal models. Many of these disorders, including obesity and auto-immune diseases, have been associated with decreased gut barrier function and consequent increased inflammation. Since A. muciniphila was found to normalize these changes and strengthen the gut barrier function, it is hypothesized that other beneficial effects of A. muciniphila might be caused by this restoration. In search for A. muciniphila's mode of action in enhancing the gut barrier function and promoting health, we reasoned that secreted components or cell envelope components of A. muciniphila are interesting candidates as they can potentially reach and interact with the epithelial barrier. In this review, we focus on the potential mechanisms through which A. muciniphila can exert its beneficial effects on the host by the production of extracellular and secreted proteins, metabolites and cell envelope components. These products have been studied in isolation for their structure, signaling capacity, and in some cases, also for their effects in preclinical models. This includes the protein known as Amuc_1100, which we here rename as pilus-associated signaling (PAS) protein , the P9 protein encoded by Amuc_1631, the short-chain fatty acids acetate and propionate, and cell envelope components, such as phosphatidylethanolamine and peptidoglycan.
Collapse
Affiliation(s)
- Anneleen Segers
- Laboratory of Microbiology, Wageningen University & Research, Wageningen 6708 WE, The Netherlands
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University & Research, Wageningen 6708 WE, The Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| |
Collapse
|
40
|
Basal Diet Fed to Recipient Mice Was the Driving Factor for Colitis and Colon Tumorigenesis, despite Fecal Microbiota Transfer from Mice with Severe or Mild Disease. Nutrients 2023; 15:nu15061338. [PMID: 36986068 PMCID: PMC10052649 DOI: 10.3390/nu15061338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Consumption of the total Western diet (TWD) in mice has been shown to increase gut inflammation, promote colon tumorigenesis, and alter fecal microbiome composition when compared to mice fed a healthy diet, i.e., AIN93G (AIN). However, it is unclear whether the gut microbiome contributes directly to colitis-associated CRC in this model. The objective of this study was to determine whether dynamic fecal microbiota transfer (FMT) from donor mice fed either the AIN basal diet or the TWD would alter colitis symptoms or colitis-associated CRC in recipient mice, which were fed either the AIN diet or the TWD, using a 2 × 2 factorial experiment design. Time-matched FMT from the donor mice fed the TWD did not significantly enhance symptoms of colitis, colon epithelial inflammation, mucosal injury, or colon tumor burden in the recipient mice fed the AIN diet. Conversely, FMT from the AIN-fed donors did not impart a protective effect on the recipient mice fed the TWD. Likewise, the composition of fecal microbiomes of the recipient mice was also affected to a much greater extent by the diet they consumed than by the source of FMT. In summary, FMT from the donor mice fed either basal diet with differing colitis or tumor outcomes did not shift colitis symptoms or colon tumorigenesis in the recipient mice, regardless of the basal diet they consumed. These observations suggest that the gut microbiome may not contribute directly to the development of disease in this animal model.
Collapse
|
41
|
Tanelian A, Nankova B, Cheriyan A, Arens C, Hu F, Sabban EL. Differences in gut microbiota associated with stress resilience and susceptibility to single prolonged stress in female rodents. Neurobiol Stress 2023; 24:100533. [PMID: 36970450 PMCID: PMC10034505 DOI: 10.1016/j.ynstr.2023.100533] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Exposure to traumatic stress is a major risk factor for the development of neuropsychiatric disorders in a subpopulation of individuals, whereas others remain resilient. The determinants of resilience and susceptibility remain unclear. Here, we aimed to characterize the microbial, immunological, and molecular differences between stress-susceptible and stress-resilient female rats before and after exposure to a traumatic experience. Animals were randomly divided into unstressed controls (n = 10) and experimental groups (n = 16) exposed to Single Prolonged Stress (SPS), an animal model of PTSD. Fourteen days later, all rats underwent a battery of behavioral tests and were sacrificed the following day to collect different organs. Stool samples were collected before and after SPS. Behavioral analyses revealed divergent responses to SPS. The SPS treated animals were further subdivided into SPS-resilient (SPS-R) and SPS-susceptible (SPS-S) subgroups. Comparative analysis of fecal 16S sequencing before and after SPS exposure indicated significant differences in the gut microbial composition, functionality, and metabolites of the SPS-R and SPS-S subgroups. In line with the observed distinct behavioral phenotypes, the SPS-S subgroup displayed higher blood-brain barrier permeability and neuroinflammation relative to the SPS-R and/or controls. These results indicate, for the first time, pre-existing and trauma-induced differences in the gut microbial composition and functionality of female rats that relate to their ability to cope with traumatic stress. Further characterization of these factors will be crucial for understanding susceptibility and fostering resilience, especially in females, who are more likely than males to develop mood disorders.
Collapse
|
42
|
Hole MJ, Jørgensen KK, Holm K, Braadland PR, Meyer‐Myklestad MH, Medhus AW, Reikvam DH, Götz A, Grzyb K, Boberg KM, Karlsen TH, Kummen M, Hov JR. A shared mucosal gut microbiota signature in primary sclerosing cholangitis before and after liver transplantation. Hepatology 2023; 77:715-728. [PMID: 36056902 PMCID: PMC9936983 DOI: 10.1002/hep.32773] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Several characteristic features of the fecal microbiota have been described in primary sclerosing cholangitis (PSC), whereas data on mucosal microbiota are less consistent. We aimed to use a large colonoscopy cohort to investigate key knowledge gaps, including the role of gut microbiota in PSC with inflammatory bowel disease (IBD), the effect of liver transplantation (LT), and whether recurrent PSC (rPSC) may be used to define consistent microbiota features in PSC irrespective of LT. APPROACH AND RESULTS We included 84 PSC and 51 liver transplanted PSC patients (PSC-LT) and 40 healthy controls (HCs) and performed sequencing of the 16S ribosomal RNA gene (V3-V4) from ileocolonic biopsies. Intraindividual microbial diversity was reduced in both PSC and PSC-LT versus HCs. An expansion of Proteobacteria was more pronounced in PSC-LT (up to 19% relative abundance) than in PSC (up to 11%) and HCs (up to 8%; Q FDR < 0.05). When investigating PSC before (PSC vs. HC) and after LT (rPSC vs. no-rPSC), increased variability (dispersion) in the PSC group was found. Five genera were associated with PSC before and after LT. A dysbiosis index calculated from the five genera, and the presence of the potential pathobiont, Klebsiella , were associated with reduced LT-free survival. Concomitant IBD was associated with reduced Akkermansia . CONCLUSIONS Consistent mucosal microbiota features associated with PSC, PSC-IBD, and disease severity, irrespective of LT status, highlight the usefulness of investigating PSC and rPSC in parallel, and suggest that the impact of gut microbiota on posttransplant liver health should be investigated further.
Collapse
Affiliation(s)
- Mikal Jacob Hole
- Norwegian PSC Research Centre, Department of Transplantation Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Kristin Kaasen Jørgensen
- Norwegian PSC Research Centre, Department of Transplantation Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Kristian Holm
- Norwegian PSC Research Centre, Department of Transplantation Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Peder R. Braadland
- Norwegian PSC Research Centre, Department of Transplantation Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Malin Holm Meyer‐Myklestad
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Ullevål, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Asle Wilhelm Medhus
- Department of Gastroenterology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Dag Henrik Reikvam
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Alexandra Götz
- Norwegian PSC Research Centre, Department of Transplantation Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Krzysztof Grzyb
- Division of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Kirsten Muri Boberg
- Norwegian PSC Research Centre, Department of Transplantation Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Tom Hemming Karlsen
- Norwegian PSC Research Centre, Department of Transplantation Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Martin Kummen
- Norwegian PSC Research Centre, Department of Transplantation Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Johannes R. Hov
- Norwegian PSC Research Centre, Department of Transplantation Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
43
|
Rausch P, Ellul S, Pisani A, Bang C, Tabone T, Marantidis Cordina C, Zahra G, Franke A, Ellul P. Microbial Dynamics in Newly Diagnosed and Treatment Naïve IBD Patients in the Mediterranean. Inflamm Bowel Dis 2023:7025776. [PMID: 36735955 DOI: 10.1093/ibd/izad004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Microbial communities have long been suspected to influence inflammatory processes in the gastrointestinal tract of patients with inflammatory bowel disease. However, these effects are often influenced by treatments and can rarely be analyzed in treatment-naïve onset cases. Specifically, microbial differences between IBD pathologies in new onset cases have rarely been investigated and can provide novel insight into the dynamics of the microbiota in Crohn's disease (CD) and ulcerative colitis (UC). METHODS Fifty-six treatment-naïve IBD onset patients (67.3% CD, 32.7% UC) and 97 healthy controls were recruited from the Maltese population. Stool samples were collected after diagnosis but before administration of anti-inflammatory treatments. Fecal microbial communities were assessed via 16S rRNA gene sequencing and subjected to ecological analyses to determine disease-specific differences between pathologies and disease subtypes or to predict future treatment options. RESULTS We identified significant differences in community composition, variability, and diversity between healthy and diseased individuals-but only small to no differences between the newly diagnosed, treatment-naïve UC and CD cohorts. Network analyses revealed massive turnover of bacterial interactions between healthy and diseased communities, as well as between CD and UC communities, as signs of disease-specific changes of community dynamics. Furthermore, we identified taxa and community characteristics serving as predictors for prospective treatments. CONCLUSION Untreated and newly diagnosed IBD shows clear differences from healthy microbial communities and an elevated level of disturbance, but only the network perspective revealed differences between pathologies. Furthermore, future IBD treatment is to some extent predictable by microbial community characteristics.
Collapse
Affiliation(s)
- Philipp Rausch
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany.,Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Sarah Ellul
- Division of Pediatric Surgery, Department of Surgery, Mater Dei Hospital, Malta
| | - Anthea Pisani
- Division of Gastroenterology, Department of Medicine, Mater Dei Hospital, Malta
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Trevor Tabone
- Division of Gastroenterology, Department of Medicine, Mater Dei Hospital, Malta
| | | | - Graziella Zahra
- Molecular Diagnostics, Department of Pathology, Mater Dei Hospital, Malta
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Pierre Ellul
- Division of Gastroenterology, Department of Medicine, Mater Dei Hospital, Malta
| |
Collapse
|
44
|
Chen J, Xiao Y, Li D, Zhang S, Wu Y, Zhang Q, Bai W. New insights into the mechanisms of high-fat diet mediated gut microbiota in chronic diseases. IMETA 2023; 2:e69. [PMID: 38868334 PMCID: PMC10989969 DOI: 10.1002/imt2.69] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/13/2022] [Accepted: 11/06/2022] [Indexed: 06/14/2024]
Abstract
High-fat diet (HFD) has been recognized as a primary factor in the risk of chronic disease. Obesity, diabetes, gastrointestinal diseases, neurodegenerative diseases, and cardiovascular diseases have long been known as chronic diseases with high worldwide incidence. In this review, the influences of gut microbiota and their corresponding bacterial metabolites on the mechanisms of HFD-induced chronic diseases are systematically summarized. Gut microbiota imbalance is also known to increase susceptibility to diseases. Several studies have proven that HFD has a negative impact on gut microbiota, also exacerbating the course of many chronic diseases through increased populations of Erysipelotrichaceae, facultative anaerobic bacteria, and opportunistic pathogens. Since bile acids, lipopolysaccharide, short-chain fatty acids, and trimethylamine N-oxide have long been known as common features of bacterial metabolites, we will explore the possibility of synergistic mechanisms among those metabolites and gut microbiota in the context of HFD-induced chronic diseases. Recent literature concerning the mechanistic actions of HFD-mediated gut microbiota have been collected from PubMed, Google Scholar, and Scopus. The aim of this review is to provide new insights into those mechanisms and to point out the potential biomarkers of HFD-mediated gut microbiota.
Collapse
Affiliation(s)
- Jiali Chen
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid DetectionJinan UniversityGuangzhouChina
- School of Chinese Medicine, Centre for Cancer and Inflammation ResearchHong Kong Baptist UniversityHong KongChina
| | - Yuhang Xiao
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid DetectionJinan UniversityGuangzhouChina
| | - Dongmei Li
- Department of Microbiology & ImmunologyGeorgetown University Medical CenterWashingtonDistrict of ColumbiaUSA
| | - Shiqing Zhang
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of PharmacyJinan UniversityGuangzhouChina
| | - Yingzi Wu
- School of Chinese Medicine, Centre for Cancer and Inflammation ResearchHong Kong Baptist UniversityHong KongChina
| | - Qing Zhang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid DetectionJinan UniversityGuangzhouChina
| | - Weibin Bai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid DetectionJinan UniversityGuangzhouChina
| |
Collapse
|
45
|
Pantoja-Feliciano IG, Karl JP, Perisin M, Doherty LA, McClung HL, Armstrong NJ, Renberg R, Racicot K, Branck T, Arcidiacono S, Soares JW. In vitro gut microbiome response to carbohydrate supplementation is acutely affected by a sudden change in diet. BMC Microbiol 2023; 23:32. [PMID: 36707764 PMCID: PMC9883884 DOI: 10.1186/s12866-023-02776-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 01/16/2023] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Interactions between diet, stress and the gut microbiome are of interest as a means to modulate health and performance. Here, in vitro fermentation was used to explore the effects of a sudden change in diet, 21 days sole sustenance on the Meal, Ready-to-Eat (MRE) U.S. military combat ration, on inter-species competition and functional potential of the human gut microbiota. Human fecal samples collected before and after MRE intervention or consuming a habitual diet (HAB) were introduced to nutrient-rich media supplemented with starch for in vitro fermentation under ascending colon conditions. 16S rRNA amplicon and Whole-metagenome sequencing (WMS) were used to measure community composition and functional potential. Specific statistical analyses were implemented to detect changes in relative abundance from taxa, genes and pathways. RESULTS Differential changes in relative abundance of 11 taxa, Dorea, Lachnospira, Bacteroides fragilis, Akkermansia muciniphila, Bifidobacterium adolescentis, Betaproteobacteria, Enterobacteriaceae, Bacteroides egerthii, Ruminococcus bromii, Prevotella, and Slackia, and nine Carbohydrate-Active Enzymes, specifically GH13_14, over the 24 h fermentation were observed as a function of the diet intervention and correlated to specific taxa of interest. CONCLUSIONS These findings suggest that consuming MRE for 21 days acutely effects changes in gut microbiota structure in response to carbohydrate but may induce alterations in metabolic capacity. Additionally, these findings demonstrate the potential of starch as a candidate supplemental strategy to functionally modulate specific gut commensals during stress-induced states.
Collapse
Affiliation(s)
| | - J. Philip Karl
- grid.420094.b0000 0000 9341 8465Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine (USARIEM), Natick, MA USA
| | - Matthew Perisin
- grid.420282.e0000 0001 2151 958XU.S. Army DEVCOM Army Research Laboratory, Adelphi, MD USA
| | - Laurel A. Doherty
- Soldier Effectiveness Directorate (SED), U.S. Army DEVCOM Soldier Center, Natick, MA USA
| | - Holly L. McClung
- grid.420094.b0000 0000 9341 8465Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine (USARIEM), Natick, MA USA
| | - Nicholes J. Armstrong
- grid.420094.b0000 0000 9341 8465Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine (USARIEM), Natick, MA USA
| | - Rebecca Renberg
- grid.420282.e0000 0001 2151 958XGeneral Technical Services, U.S. Army DEVCOM Army Research Laboratory, Adelphi, MD USA
| | - Kenneth Racicot
- Soldier Effectiveness Directorate (SED), U.S. Army DEVCOM Soldier Center, Natick, MA USA
| | - Tobyn Branck
- Soldier Effectiveness Directorate (SED), U.S. Army DEVCOM Soldier Center, Natick, MA USA
| | - Steve Arcidiacono
- Soldier Effectiveness Directorate (SED), U.S. Army DEVCOM Soldier Center, Natick, MA USA
| | - Jason W. Soares
- Soldier Effectiveness Directorate (SED), U.S. Army DEVCOM Soldier Center, Natick, MA USA
| |
Collapse
|
46
|
Reznikov EA, Suskind DL. Current Nutritional Therapies in Inflammatory Bowel Disease: Improving Clinical Remission Rates and Sustainability of Long-Term Dietary Therapies. Nutrients 2023; 15:nu15030668. [PMID: 36771373 PMCID: PMC9920576 DOI: 10.3390/nu15030668] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/19/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Inflammatory Bowel Disease (IBD) includes a spectrum of chronic immune-mediated intestinal diseases thought to be related to the complex interaction between the host immune system and the intestinal microbiome. Research supports the use of nutritional therapy in IBD; however, it is not routinely used in clinical practice. This literature review seeks to advance the understanding of diet and its effect in IBD with a focus on both Crohn's Disease (CD) and Ulcerative Colitis (UC). The contribution of diet to the development and treatment of IBD cannot be overstated. In both pediatric as well as adult IBD, nutritional interventions have been shown to improve clinical symptoms as well as inflammatory burden. The impact of dietary intervention is best exemplified through the use of Exclusive Enteral Nutrition (EEN) in CD. EEN and clinical research on exclusionary whole food diets-Crohn's Disease Exclusion Diet (CDED), Specific Carbohydrate Diet (SCD), low fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAP) diet, and Mediterranean Diet-are discussed within this review. Current clinical literature supports the elimination of detrimental components and the incorporation of low processed whole foods in the diet. Additional prospective and longitudinal dietary studies on sustainable and long-term dietary options, along with a deeper understanding of the mechanism, are needed to further advance the role of nutritional interventions in IBD.
Collapse
|
47
|
Heo M, Park YS, Yoon H, Kim NE, Kim K, Shin CM, Kim N, Lee DH. Potential of Gut Microbe-Derived Extracellular Vesicles to Differentiate Inflammatory Bowel Disease Patients from Healthy Controls. Gut Liver 2023; 17:108-118. [PMID: 36424722 PMCID: PMC9840915 DOI: 10.5009/gnl220081] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/09/2022] [Accepted: 06/03/2022] [Indexed: 11/26/2022] Open
Abstract
Background/Aims This study aimed to evaluate the potential of the stool microbiome and gut microbe-derived extracellular vesicles (EVs) to differentiate between patients with inflammatory bowel disease (IBD) and healthy controls, and to predict relapse in patients with IBD. Methods Metagenomic profiling of the microbiome and bacterial EVs in stool samples of controls (n=110) and patients with IBD (n=110) was performed using 16S rRNA sequencing and then compared. Patients with IBD were divided into two enterotypes based on their microbiome, and the cumulative risk of relapse was evaluated. Results There was a significant difference in the composition of the stool microbiome and gut microbe-derived EVs between patients with IBD and controls. The alpha diversity of the microbiome in patients with IBD was significantly lower than that in controls, while the beta diversity also differed significantly between the two groups. These findings were more prominent in gut microbe-derived EVs than in the stool microbiome. The survival curve tended to be different for enterotypes based on the gut microbe-derived EVs; however, this difference was not statistically significant (log-rank test, p=0.166). In the multivariable analysis, elevated fecal calprotectin (>250 mg/kg) was the only significant risk factor associated with relapse (adjusted hazard ratio, 3.147; 95% confidence interval, 1.545 to 6.408; p=0.002). Conclusions Analysis of gut microbe-derived EVs is better at differentiating patients with IBD from healthy controls than stool microbiome analysis.
Collapse
Affiliation(s)
- Min Heo
- Interdisciplinary Program of Bioinformatics, College of Natural Sciences, Seoul National University, Seoul, Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea,Corresponding AuthorHyuk Yoon, ORCIDhttps://orcid.org/0000-0002-2657-0349, E-mail
| | - Nam-Eun Kim
- Department of Public Health Science, Graduate School of Public Health, Seoul, Korea
| | - Kangjin Kim
- Institute of Health and Environment, Seoul National University, Seoul, Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
48
|
Zheng M, Han R, Yuan Y, Xing Y, Zhang W, Sun Z, Liu Y, Li J, Mao T. The role of Akkermansia muciniphila in inflammatory bowel disease: Current knowledge and perspectives. Front Immunol 2023; 13:1089600. [PMID: 36685588 PMCID: PMC9853388 DOI: 10.3389/fimmu.2022.1089600] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/20/2022] [Indexed: 01/08/2023] Open
Abstract
Inflammatory bowel diseases, including Crohn's disease and ulcerative colitis, is a chronic relapsing gastrointestinal inflammatory disease mediated by dysregulated immune responses to resident intestinal microbiota. Current conventional approaches including aminosalicylates, corticosteroids, immunosuppressive agents, and biological therapies are focused on reducing intestinal inflammation besides inducing and maintaining disease remission, and managing complications. However, these therapies are not curative and are associated with various limitations, such as drug resistance, low responsiveness and adverse events. Recent accumulated evidence has revealed the involvement of mucin-degrading bacterium Akkermansia muciniphila (A. muciniphila) in the regulation of host barrier function and immune response, and how reduced intestinal colonisation of probiotic A. muciniphila can contribute to the process and development of inflammatory bowel diseases, suggesting that it may be a potential target and promising strategy for the therapy of inflammatory bowel disease. In this review, we summarise the current knowledge of the role of A. muciniphila in IBD, especially focusing on the related mechanisms, as well as the strategies based on supplementation with A. muciniphila, probiotics and prebiotics, natural diets, drugs, and herbs to promote its colonisation in the gut, and holds promise for A. muciniphila-targeted and -based therapies in the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
| | - Ran Han
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yali Yuan
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yunqi Xing
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wenji Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | | | - Yuyue Liu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junxiang Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tangyou Mao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
49
|
Panah FM, Nielsen KD, Simpson GL, Schönherz A, Schramm A, Lauridsen C, Nielsen TS, Højberg O, Fredborg M, Purup S, Canibe N. A westernized diet changed the colonic bacterial composition and metabolite concentration in a dextran sulfate sodium pig model for ulcerative colitis. Front Microbiol 2023; 14:1018242. [PMID: 37138607 PMCID: PMC10150118 DOI: 10.3389/fmicb.2023.1018242] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction Ulcerative colitis (UC) is characterized by chronic inflammation in the colonic epithelium and has a blurred etiology. A western diet and microbial dysbiosis in the colon were reported to play a role in UC development. In this study, we investigated the effect of a westernized diet, i.e., increasing fat and protein content by including ground beef, on the colonic bacterial composition in a dextran sulfate sodium (DexSS) challenged pig study. Methods The experiment was carried out in three complete blocks following a 2×2 factorial design including 24 six-week old pigs, fed either a standard diet (CT) or the standard diet substituted with 15% ground beef to simulate a typical westernized diet (WD). Colitis was induced in half of the pigs on each dietary treatment by oral administration of DexSS (DSS and WD+DSS, respectively). Samples from proximal and distal colon and feces were collected. Results and discussion Bacterial alpha diversity was unaffected by experimental block, and sample type. In proximal colon, WD group had similar alpha diversity to CT group and the WD+DSS group showed the lowest alpha diversity compared to the other treatment groups. There was a significant interaction between western diet and DexSS for beta diversity, based on Bray-Curtis dissimilarly. The westernized diet and DexSS resulted in three and seven differentially abundant phyla, 21 and 65 species, respectively, mainly associated with the Firmicutes and Bacteroidota phyla followed by Spirochaetota, Desulfobacterota, and Proteobacteria. The concentration of short-chain fatty acids (SCFA) was lowest in the distal colon. Treatment had a slight effect on the estimates for microbial metabolites that might have valuable biological relevance for future studies. The concentration of putrescine in the colon and feces and that of total biogenic amines was highest in the WD+DSS group. We conclude that a westernized diet could be a potential risk factor and an exacerbating agent for UC by reducing the abundance of SCFA-producing bacteria, increasing the abundance of pathogens such as Helicobacter trogontum, and by increasing the concentration of microbial proteolytic-derived metabolites in the colon.
Collapse
Affiliation(s)
- Farhad M. Panah
- Department of Veterinary and Animal Sciences, Aarhus University, Tjele, Denmark
- *Correspondence: Farhad M. Panah,
| | - Katrine D. Nielsen
- Department of Veterinary and Animal Sciences, Aarhus University, Tjele, Denmark
| | - Gavin L. Simpson
- Department of Veterinary and Animal Sciences, Aarhus University, Tjele, Denmark
| | - Anna Schönherz
- Department of Veterinary and Animal Sciences, Aarhus University, Tjele, Denmark
| | | | - Charlotte Lauridsen
- Department of Veterinary and Animal Sciences, Aarhus University, Tjele, Denmark
| | - Tina S. Nielsen
- Department of Veterinary and Animal Sciences, Aarhus University, Tjele, Denmark
| | - Ole Højberg
- Department of Veterinary and Animal Sciences, Aarhus University, Tjele, Denmark
| | - Marlene Fredborg
- Department of Veterinary and Animal Sciences, Aarhus University, Tjele, Denmark
| | - Stig Purup
- Department of Veterinary and Animal Sciences, Aarhus University, Tjele, Denmark
| | - Nuria Canibe
- Department of Veterinary and Animal Sciences, Aarhus University, Tjele, Denmark
| |
Collapse
|
50
|
Dai XC, Zhang YH, Huang YL, Wu XT, Fang YJ, Gao YJ, Wang F. Calorie restriction remodels gut microbiota and suppresses tumorigenesis of colorectal cancer in mice. Exp Ther Med 2022; 25:59. [PMID: 36588818 PMCID: PMC9780522 DOI: 10.3892/etm.2022.11758] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide and the consumption of a high-calorie diet is one of its risk factors. Calorie restriction (CR) slows tumor growth in a variety of cancers, including colorectal cancer; however, the mechanism behind this remains unknown. In the present study, CR effectively reduced the tumor volume and weight in a xenograft BALB/c male nude mouse model. In addition, tumor immunohistochemistry revealed that the CR group had significantly higher expression of Bax (P<0.001) and significantly lower levels of Bcl2 (P<0.0001) and Ki67 (P<0.001) compared with control group. Furthermore, data from 16S ribosomal (r)RNA sequencing implied that CR was able to reprogram the microbiota structure, characterized by increased Lactobacillus constituent ratio (P<0.05), with amelioration of microbial dysbiosis caused by CRC. Further receiver operating characteristic curves demonstrated that the bacteria Bacteroides [area under the curve (AUC)=0.800], Lactobacillus (AUC=0.760) and Roseburia (AUC=0.720) served key roles in suppression of CRC in the mouse model. The functional prediction of intestinal flora indicated 'cyanoamino acid metabolism' (P<0.01), 'replication initiation protein REP (rolling circle plasmid replication)' (P<0.01), 'tRNA G10 N-methylase Trm11' (P<0.01) and 'uncharacterized protein with cyclophilin fold, contains DUF369 domain' (P<0.05) were downregulated in CR group. These findings implied that CR suppressed CRC in mice and altered the gut microbiota.
Collapse
Affiliation(s)
- Xing-Chen Dai
- Department of Gastroenterology, General Hospital, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China,School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yu-Huan Zhang
- Department of Gastroenterology, General Hospital, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China,School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yong-Li Huang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xiao-Ting Wu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yu-Jie Fang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yu-Jing Gao
- Department of Gastroenterology, General Hospital, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China,National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China,Correspondence to: Professor Yu-Jing Gao, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, P.R. China
| | - Fang Wang
- Department of Gastroenterology, General Hospital, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China,Correspondence to: Professor Yu-Jing Gao, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|