1
|
Yan H, Hua Y, Ni J, Wu X, Xu J, Zhang Z, Dong J, Xiong Z, Yang L, Yuan H. Acupuncture ameliorates inflammation by regulating gut microbiota in acute ischemic stroke. IBRO Neurosci Rep 2025; 18:443-452. [PMID: 40144797 PMCID: PMC11938260 DOI: 10.1016/j.ibneur.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/15/2025] [Indexed: 03/28/2025] Open
Abstract
Background Acute ischemic stroke(AIS) is a major life-threatening disease.Some studies have found that AIS may be related to gut flora and immune responses. Acupuncture is used widely in the treatment of AIS. However its relevant mechanism is unclear enough. Therefore, in this study, we wanted to confirm that acupuncture was treating AIS through gut flora and immune response. Methods We randomly divided 18 rats into equal three groups, including Sham, Middle Cerebral Artery Occlusion (MCAO) and Acupuncture.Rats in the Acupuncture group for a continuous period of three days after surgery. Neurological deficits were assessed using Longa's method, and detection of intestinal flora by 16s rRNA gene sequencing, determination of SCFAs by gas chromatography-mass spectrometry, detection of HDAC and inflammatory cytokines by elisa assay, detection of Th17 and Treg cells by flow cytometry and, observation of pathological and morphological changes in brain and colon tissues by HE staining. Results Acupuncture improved the degree of impaired neurological function in MCAO rats and regulated the type and abundance of intestinal bacteria, increased SCFAs of MCAO rats, decreased HDAC1 and HDAC2, modulated the Th17/Treg imbalance, reduced the level of inflammatory factors in the peripheral blood and altered the pathology of the intestine and brain. Conclusion Acupuncture repaired neurologic deficits after AIS and may be associated with an immune-inflammatory response mediated by gut microbiota.
Collapse
Affiliation(s)
- Haoyue Yan
- Department of Acupuncture and Moxibustion, Dongzhimen Hospital of Beijing University of Chinese Medicine, China
| | - Yini Hua
- Department of Acupuncture and Moxibustion, Dongzhimen Hospital of Beijing University of Chinese Medicine, China
| | - Jinxia Ni
- Department of Acupuncture and Moxibustion, Dongzhimen Hospital of Beijing University of Chinese Medicine, China
| | - Xiaona Wu
- Department of Rehabilitation, Beijing Fengtai Hospital, China
| | - Jingni Xu
- Dongcheng District Dongzhimen Community Healthcare Center, China
| | - Ziniu Zhang
- Department of Acupuncture and Moxibustion, Dongzhimen Hospital of Beijing University of Chinese Medicine, China
| | - Juwei Dong
- Department of Acupuncture and Moxibustion, Dongzhimen Hospital of Beijing University of Chinese Medicine, China
| | - Zhihao Xiong
- Department of Acupuncture and Moxibustion, Dongzhimen Hospital of Beijing University of Chinese Medicine, China
| | - Lei Yang
- Department of Acupuncture and Moxibustion, Dongzhimen Hospital of Beijing University of Chinese Medicine, China
| | - Hongwei Yuan
- Department of Acupuncture and Moxibustion, Dongzhimen Hospital of Beijing University of Chinese Medicine, China
| |
Collapse
|
2
|
Overton EN, Zhang Y, Ngecu W, Seyedsayamdost MR. Chemical Synthetic Lethality Screens Identify Selective Drug Combinations against Pseudomonas aeruginosa. ACS Chem Biol 2025. [PMID: 40258132 DOI: 10.1021/acschembio.5c00076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
The emergence of bacterial ESKAPE pathogens presents a formidable challenge to global health, necessitating the development of innovative strategies for antibiotic discovery. Here, we leverage chemical synthetic lethality to locate therapeutic combinations of small molecules against multidrug-resistant Pseudomonas aeruginosa. Using a transposon screen, we identify PyrD as a target for sensitizing P. aeruginosa to subinhibitory doses of ceftazidime. High-throughput inhibitor screens identify two PyrD inhibitors, nordihydroguaiaretic acid (NDGA) and chlorhexidine (CHX), each of which does not significantly affect growth in isolation but exhibits chemical synthetic lethality when combined with low-dose ceftazidime. Downstream biochemical studies elucidate the mechanism of inhibition by NDGA and CHX. Remarkably, this combination is toxic to P. aeruginosa but leaves commensal bacteria, which are more susceptible to antibiotics, unscathed. Aside from advancing drug combinations that may be explored further in the future, our results offer a new approach for devising potent and specific drug combinations against recalcitrant pathogens.
Collapse
Affiliation(s)
- Ellysia N Overton
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Yifan Zhang
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
| | | | - Mohammad R Seyedsayamdost
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
3
|
Beyoğlu D, Idle JR. The Microbiome and Metabolic Dysfunction-Associated Steatotic Liver Disease. Int J Mol Sci 2025; 26:2882. [PMID: 40243472 PMCID: PMC11988851 DOI: 10.3390/ijms26072882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a condition wherein excessive fat accumulates in the liver, leading to inflammation and potential liver damage. In this narrative review, we evaluate the tissue microbiota, how they arise and their constituent microbes, and the role of the intestinal and hepatic microbiota in MASLD. The history of bacteriophages (phages) and their occurrence in the microbiota, their part in the potential causation of MASLD, and conversely, "phage therapy" for antibiotic resistance, obesity, and MASLD, are all described. The microbiota metabolism of bile acids and dietary tryptophan and histidine is defined, together with the impacts of their individual metabolites on MASLD pathogenesis. Both periodontitis and intestinal microbiota dysbiosis may cause MASLD, and how individual microorganisms and their metabolites are involved in these processes is discussed. Novel treatment opportunities for MASLD involving the microbiota exist and include fecal microbiota transplantation, probiotics, prebiotics, synbiotics, tryptophan dietary supplements, intermittent fasting, and phages or their holins and endolysins. Although FDA is yet to approve phage therapy in clinical use, there are multiple FDA-approved clinical trials, and this may represent a new horizon for the future treatment of MASLD.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
| | - Jeffrey R. Idle
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
- Department of Biomedical Research, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
4
|
Westmark CJ. Soy-based purified ingredient diet affects mouse gut permeability and the microbiome in fragile X mice. Front Mol Neurosci 2025; 18:1520211. [PMID: 40190341 PMCID: PMC11968763 DOI: 10.3389/fnmol.2025.1520211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/28/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Gastrointestinal problems including vomiting, reflux, flatulence, diarrhea, constipation and colic are common comorbidities in fragile X syndrome. There is accumulating evidence suggesting that leaky gut syndrome causes neurological phenotypes. Although fragile X messenger ribonucleoprotein is ubiquitously expressed, there is a dearth of knowledge regarding its role outside of the brain including effects on gut dysfunction in fragile X. The aim of this study was to generate novel data on gastrointestinal barrier function and the gut microbiome in response to Fmr1 genotype, sex and diet in mice. Methods Fmr1KO male mice and littermate controls in an FVB background were maintained on two purified ingredient diets (AIN-93G with casein protein versus soy protein isolate) versus two standard chows (Teklad 2019 with wheat, corn and yeast protein versus Purina 5015 with wheat, soy, corn, yeast and whey protein sources). Gut permeability was quantified by FITC-dextran levels in blood plasma. The cecal microbiome was identified by 16S rRNA sequencing. In addition, gut permeability was tested in Fmr1KO mice in the C57BL/6 J background maintained on casein- and soy protein isolate-based AIN-93G versus Teklad 2019. Results Knockout of the Fmr1 gene in FVB mice did not affect gut permeability. Soy protein isolate-based AIN-93G increased gut permeability. Beta-diversity of the cecal microbiome was significantly altered as a function of the four test diets. Akkermansia_muciniphila was increased in Fmr1KO mice fed AIN-93G while unnamed species within the genus Anaerovorax and family Ruminococcaceae were increased and the order Clostridales decreased in Fmr1KO mice fed AIN-93G/soy. Fmr1KO mice in the C57BL/6 J background exhibited increased gut permeability in response to soy protein. Discussion These findings regarding the effects of diet on gut permeability and the microbiome have important implications for experimental design. Single-source diets are ubiquitously used to maintain laboratory animals for medical research and feed details are frequently not reported in publications. Diet/phenotype interactions could have a large impact on inter-laboratory replicability in premedical research. For infants with fragile X, early-life diet could impact the severity of disease outcomes.
Collapse
Affiliation(s)
- Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI, United States
- Molecular Environmental Toxicology Center, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
5
|
Cortijo-Alfonso ME, Laghouaouta H, Pena RN, Martínez M, Yuste S, Rubió-Piqué L, Piñol-Felis C. Gut microbiota modulation and inflammation mitigation in a murine model through a hull-less and purple grain barley genotype. Food Funct 2025; 16:2389-2400. [PMID: 39996301 DOI: 10.1039/d4fo05524a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Barley, increasingly recognized for its health benefits, contains bioactive compounds like beta-glucans and (poly)phenols. Newly developed purple barley varieties, enriched with anthocyanins, offer potential gut health benefits. This study examined the effects of a hull-less, purple-grain barley genotype, consumed as whole-grain or isolated fractions (bran and endosperm), on gut microbiota and inflammation in a murine model. Fifty male and female BALB/cB&J mice were assigned to five diets over six weeks: standard diet (SD), rice diet (RD), whole-grain barley (WGB), anthocyanin-rich barley bran (BB), and beta-glucan-rich endosperm (PG). The BB diet triggered anti-inflammatory signals as it reduced IFN-γ and IL-4 in females, lowered TNF-α in both sexes, and decreased C-Reactive Protein (CRP) in males compared to SD. The PG diet improved gut barrier integrity by lowering LPS-binding protein levels. Barley-based diets enhanced gut microbiota diversity, particularly, by increasing beneficial bacteria like Lactobacillus, Lachnospiraceae UCG-001, and Akkermansia. Notably, BB and PG elicited stronger effects than WGB, suggesting that grain fractionation modifies the food matrix, potentially enhancing the bioaccessibility and bioavailability of key bioactive compounds. These results underscore the benefits of purple barley-derived fractions in promoting gut health and reducing inflammation, supporting their potential role to protect against inflammation-related conditions.
Collapse
Affiliation(s)
| | - Houda Laghouaouta
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 191 Rovira Roure, 25198, Lleida, Catalonia, Spain
| | - Ramona N Pena
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 191 Rovira Roure, 25198, Lleida, Catalonia, Spain
| | - Mariona Martínez
- University of Lleida-Agrotecnio CERCA Center, Av. Alcalde Rovira Roure 191, 25198 Lleida, Spain.
| | - Silvia Yuste
- University of Lleida-Agrotecnio CERCA Center, Av. Alcalde Rovira Roure 191, 25198 Lleida, Spain.
| | - Laura Rubió-Piqué
- University of Lleida-Agrotecnio CERCA Center, Av. Alcalde Rovira Roure 191, 25198 Lleida, Spain.
| | - Carme Piñol-Felis
- Department of Medicine and Surgery, University of Lleida, Lleida, Catalonia, Spain
- Institut de Recerca Biomèdica de Lleida, Fundació Dr Pifarré IRBLleida, Lleida, Catalonia, Spain
| |
Collapse
|
6
|
Ran R, Uslu M, Siddiqui MF, Brubaker DK, Trapecar M. Single-Cell Analysis Reveals Tissue-Specific T Cell Adaptation and Clonal Distribution Across the Human Gut-Liver-Blood Axis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642626. [PMID: 40161783 PMCID: PMC11952442 DOI: 10.1101/2025.03.11.642626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Understanding T cell clonal relationships and tissue-specific adaptations is crucial for deciphering human immune responses, particularly within the gut-liver axis. We performed paired single-cell RNA and T cell receptor sequencing on matched colon (epithelium, lamina propria), liver, and blood T cells from the same human donors. This approach tracked clones across sites and assessed microenvironmental impacts on T cell phenotype. While some clones were shared between blood and tissues, colonic intraepithelial lymphocytes (IELs) exhibited limited overlap with lamina propria T cells, suggesting a largely resident population. Furthermore, tissue-resident memory T cells (TRM) in the colon and liver displayed distinct transcriptional profiles. Notably, our analysis suggested that factors enriched in the liver microenvironment may influence the phenotype of colon lamina propria TRM. This integrated single-cell analysis maps T cell clonal distribution and adaptation across the gut-liver-blood axis, highlighting a potential liver role in shaping colonic immunity.
Collapse
Affiliation(s)
- Ran Ran
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Merve Uslu
- Department of Medicine, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Mohd Farhan Siddiqui
- Department of Medicine, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Douglas K. Brubaker
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH
- The Blood, Heart, Lung, and Immunology Research Center, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, OH
| | - Martin Trapecar
- Department of Medicine, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
7
|
Li S, Wang H, Li B, Lu H, Zhao J, Gao A, An Y, Yang J, Ma T. Multi-Omics Analysis Reveals the Negative Effects of High-Concentrate Diets on the Colonic Epithelium of Dumont Lambs. Animals (Basel) 2025; 15:749. [PMID: 40076032 PMCID: PMC11898968 DOI: 10.3390/ani15050749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Feeding HC diets has been found to induce metabolic dysregulation in the colon. However, the mechanisms by which changes in colonic flora and metabolites damage the colonic epithelium are poorly studied. Therefore, the present experiment used a multi-omics technique to investigate the mechanism of colonic injury induced by high-concentrate diets in lambs. Twelve male Dumont lambs were randomly split into two groups: a low-concentrate diet (LC = concentrate/forage = 30:70) group and a high-concentrate diet (HC = concentrate/forage = 70:30) group. The results showed that the HC group presented significantly increased lipopolysaccharide (LPS) concentrations in the colonic epithelium and significantly decreased serum total cholesterol (TC), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) levels (p < 0.05), which led to cavities and inflammatory cell infiltration in the colonic epithelium. The HC group had significantly lower pH and less VFAs in colon contents, as well as a significantly increased abundance of bacteria of the genera [Eubacterium]_coprostanoligenes_group, Rikenellaceae_RC9_gut_group, Treponema, Clostridia_UCG-014, Alistipes, Ruminococcus, Christensenellaceae_R-7_group, UCG-002, Bacteroidales_RF16_group and Lachnospiraceae_AC2044_group compared to the LC diet group. These microorganisms significantly increased the level of metabolites of cholic acid, chenodeoxycholic acid, LysoPA (P-16:0/0:0), methapyrilene, and fusaric acid. A transcriptome analysis showed that cytokine-cytokine receptor interaction, glutathione metabolism, and the peroxisome signaling pathway were downregulated in the colon epithelium of the lambs fed the HC diet. Therefore, the HC diet caused epithelial inflammation and oxidative damage by affecting the interaction between the microbial flora of the colon and metabolites and the host epithelium, which eventually disrupted colon homeostasis and had a negative impact on sheep health.
Collapse
Affiliation(s)
- Shufang Li
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| | - Hairong Wang
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| | - Boyang Li
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| | - Henan Lu
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| | - Jianxin Zhao
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| | - Aiwu Gao
- Food Science, Inner Mongolia Agricultural University, Hohhot 010018, China;
| | - Yawen An
- Veterinary Research Institute, Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot 010018, China;
| | - Jinli Yang
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| | - Tian Ma
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| |
Collapse
|
8
|
He ZH, Jin Y, Chen D, Zheng HX, Xiang JE, Jiang YJ, Wen ZS. Seleno-chitooligosaccharide-induced modulation of intestinal barrier function: Role of inflammatory cytokines, tight junction proteins, and gut microbiota in mice. J Appl Biomed 2025; 23:45-55. [PMID: 40145885 DOI: 10.32725/jab.2025.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/26/2025] [Indexed: 03/28/2025] Open
Abstract
This study aimed to explore the function of Seleno-chitooligosaccharide (SOA) on the intestinal barrier through regulation of inflammatory cytokines, tight junction protein, and gut microbiota in mice. The results of ELISA assay demonstrated that SOA significantly increased the levels of IL-2, IL-10, and IFN-γ in serum and ileum. Meanwhile, SOA increased the levels of IL-4 in the ileum (p < 0.05). In addition, Diamine Oxidase (DAO) concentration was decreased in ileum by SOA treatments (p < 0.05). The administration of SOA significantly upregulated the expression of ZO-1 and Occludin in the ileum (p < 0.05). By 16S rDNA sequencing, reduced ratio of Bacillota/Bacteroidota was observed in SOA treated mice. Within the phylum of Bacteroidota, SOA increased the relative abundance of Deferribacterota, uncultured Bacteroidales bacterium, and Bacteroides. Within the phylum of Bacillota, increased relative abundance of Erysipelatoclostridium and Lachnoclostridium, and reduced relative abundance of Ruminococcaceae UCG-010 were observed with SOA supplement. In summary, SOA has the potential to modulate the function of intestinal barrier function and prevent intestinal diseases.
Collapse
Affiliation(s)
| | | | - Die Chen
- Zhejiang Ocean University, School of Food and Pharmacy, Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Zhoushan, Zhejiang Province 316022, China
| | - Hui-Xin Zheng
- Zhejiang Ocean University, School of Food and Pharmacy, Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Zhoushan, Zhejiang Province 316022, China
| | - Jia-Er Xiang
- Zhejiang Ocean University, School of Food and Pharmacy, Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Zhoushan, Zhejiang Province 316022, China
| | - Yong-Jun Jiang
- Zhejiang Ocean University, School of Food and Pharmacy, Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Zhoushan, Zhejiang Province 316022, China
| | - Zheng-Shun Wen
- Zhejiang Ocean University, School of Food and Pharmacy, Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Zhoushan, Zhejiang Province 316022, China
- Xianghu Lab, Venture Valley Building, 168 Gengwen Road, Xiaoshan District, Hangzhou, Zhejiang Province 311231, China
| |
Collapse
|
9
|
Fan X, Lv N, Quan Z. Culturable Human Microorganisms and the Impact of Transportation Conditions on Cultivability. Microorganisms 2025; 13:549. [PMID: 40142442 PMCID: PMC11944332 DOI: 10.3390/microorganisms13030549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
The composition of the human microbiome is a critical health indicator, and culture-independent methodologies have substantially advanced our understanding of human-associated microorganisms. However, precise identification and characterization of microbial strains require culture-based techniques. Recently, the resurgence of culturomics, combined with high-throughput sequencing technology, has reduced the high labor demand of pure culture methods, facilitating a more efficient and comprehensive acquisition of culturable microbial strains. This study employed an integrated approach combining culturomic and high-throughput sequencing to identify culturable microorganisms on the human scalp and in human saliva and feces. Several Staphylococcus strains were identified from the scalp, whereas anaerobic microorganisms were dominant in the saliva and fecal samples. Additionally, the study highlighted the beneficial effects of transportation conditions (liquid nitrogen treatment, dry ice transport, and dimethyl sulfoxide [DMSO] buffer) in preserving culturable microorganisms. A robust methodology was developed for the large-scale acquisition of culturable microorganisms with optimized transport conditions that enhance the potential for isolating a greater diversity of culturable strains.
Collapse
Affiliation(s)
| | | | - Zhexue Quan
- Microbiome Center, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China; (X.F.); (N.L.)
| |
Collapse
|
10
|
Adair MG, Tolley KA, van Vuuren BJ, da Silva JM. Anthropogenic reverberations on the gut microbiome of dwarf chameleons ( Bradypodion). PeerJ 2025; 13:e18811. [PMID: 40034670 PMCID: PMC11874949 DOI: 10.7717/peerj.18811] [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: 07/05/2024] [Accepted: 12/12/2024] [Indexed: 03/05/2025] Open
Abstract
Exploration of the microbiome has been referred to as a final frontier in biological research. This is due to its precedence for generating insights on the holistic functioning of organismal biology by exploring the interactions between hosts and their associated symbiotic organisms. The microbiomes of many vertebrate groups still require exploration to advance current knowledge and fill previous knowledge gaps. This study generated initial descriptions of the bacterial microbiomes of three species of dwarf chameleon (Bradypodion) from the 16S rRNA gene region targeting the V3 and V4 hypervariable regions. This led to the successful identification of 1,073 and 4,502 independent amplicon sequence variants from buccal swab and faecal material samples, respectively. This newly acquired information is intended as a baseline for future work incorporating holobiont information. The diversity of microbial taxa suggests that the total dwarf chameleon microbiome is similar to other squamates investigated to date, as well as chelonians (Testudines). Microbial frequency differences were noted in comparison to crocodilians (Archosauria) and mammalian groups. Furthermore, this study aimed to examine the influence of habitat transformation on the composition of the microbiome in dwarf chameleons as each of the study species occupy both urban and natural habitats. Given that most urban habitats are highly transformed, the expectation was that microbial assemblages of the gastro-intestinal tracts of all three Bradypodion species would show significant differences between populations (i.e., natural, or urban). It was found, however, that the level of effect was contingent on species: B. melanocephalum populations showed noticeable microbiome differences between urban and natural populations; B. thamnobates showed variations in microbial community dispersions between populations; and B. setaroi showed no significant microbiome differences based on diversity metrics although some frequency differences, in microbiome composition, were observed between populations. We suggest that the magnitude of difference between the habitats occupied by the populations is a factor, given the apparent disparity between the natural and urban habitats for B. melanocephalum as compared to the other two species.
Collapse
Affiliation(s)
- Matthew G. Adair
- Kirstenbosch Research Centre, South African National Biodiversity Institute, Cape Town, Newlands, South Africa
- Centre for Ecological Genomics and Wildlife Conservation, University of Johannesburg, Johannesburg, Gauteng, South Africa
| | - Krystal A. Tolley
- Kirstenbosch Research Centre, South African National Biodiversity Institute, Cape Town, Newlands, South Africa
- Centre for Ecological Genomics and Wildlife Conservation, University of Johannesburg, Johannesburg, Gauteng, South Africa
| | - Bettine Jansen van Vuuren
- Centre for Ecological Genomics and Wildlife Conservation, University of Johannesburg, Johannesburg, Gauteng, South Africa
| | - Jessica Marie da Silva
- Kirstenbosch Research Centre, South African National Biodiversity Institute, Cape Town, Newlands, South Africa
- Centre for Ecological Genomics and Wildlife Conservation, University of Johannesburg, Johannesburg, Gauteng, South Africa
| |
Collapse
|
11
|
Bhure M, Savaliya K, Patil S, Nehra C, Pandit R, Shah T, Patil NV, Patel AK, Kachhawaha S, Kumawat RN, Joshi M, Joshi CG. Bioprospecting of 101 facultative rumen bacterial isolates through comprehensive genome analysis. Mol Biol Rep 2025; 52:265. [PMID: 40014144 DOI: 10.1007/s11033-025-10291-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/22/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Microbes within the rumen play a pivotal role in the digestion of feed ingested by the ruminants. Researchers have been investigating microbes within rumen to assess its genetic capabilities, which hold immense potential across various fields including agro-industrial advantages. Since rumen is preliminary an anaerobic sac, numerous anaerobic bacteria and fungi have been isolated and characterized, however facultative anaerobic bacteria yet not fully investigated. METHODS AND RESULTS In present study, we isolated, characterized and performed whole genome analysis of 101 facultative anaerobic bacteria from rumen, offering a unique perspective compared to metagenomic approaches. All assembled genomes were of high quality, i.e. completeness 100% (only seven were between 92 and 99.5%) and only two had contamination > 5%. We identified 9,542 sequences of Carbohydrate-Active Enzymes (CAZymes). Over 8,136 of these CAZymes were full-length sequences, with 2,048 harbouring signal peptides also. Xylan (n = 634), pectin (n = 604), and starch (n = 312) degrading enzyme sequences were dominant. Several isolates also harbour secondary metabolite biosynthesis gene clusters for various metabolites, including fengycin, lichenysin, bacillibactins, bacilysin etc. All the isolates have metabolic versatility, encompassing pathways such as carbohydrate, amino acid, lipid, and vitamin and cofactor metabolism. Intriguingly, lipoic acid metabolism was absent in most of these facultative bacterial isolates. CONCLUSION This comprehensive study sheds light on the genetic potential of culturable facultative rumen bacteria, emphasizing their pivotal roles in carbohydrate degradation, secondary metabolite production, and metabolic diversity. These findings hold promise for enhancing ruminant nutrition, advancing eco-friendly biomass conversion, and bolstering bioprospecting of industrially important biocules and enzymes biofuel production.
Collapse
Affiliation(s)
- Minal Bhure
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, 382011, India
| | - Kaksha Savaliya
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, 382011, India
| | - Sonal Patil
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, 382011, India
| | - Chitra Nehra
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, 382011, India
| | - Ramesh Pandit
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, 382011, India
| | - Tejas Shah
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, 382011, India
| | - Niteen V Patil
- ICAR-Central Arid Zone Research Institute, Jodhpur, Rajasthan, 342005, India
| | - Ashutosh K Patel
- ICAR-Central Arid Zone Research Institute, Jodhpur, Rajasthan, 342005, India
| | - Subhash Kachhawaha
- ICAR-Central Arid Zone Research Institute, Jodhpur, Rajasthan, 342005, India
| | - Ram N Kumawat
- ICAR-Central Arid Zone Research Institute, Jodhpur, Rajasthan, 342005, India
| | - Madhvi Joshi
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, 382011, India
| | - Chaitanya G Joshi
- Gujarat Biotechnology Research Centre, Gandhinagar, Gujarat, 382011, India.
| |
Collapse
|
12
|
Dardi P, Coutinho CP, de Oliveira S, Teixeira SA, Gacek RRF, Purgatto E, Vinolo MAR, Muscará MN, Rossoni LV. Changes in the intestinal microbiota induced by the postnatal environment and their association with hypertension. Pharmacol Res 2025; 212:107621. [PMID: 39848350 DOI: 10.1016/j.phrs.2025.107621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
It has been established that cross-fostering impacts the development of hypertension in spontaneously hypertensive rats (SHR). However, the ability of the cross-fostering protocol to shape gut microbiota profile in SHR and impact hypertension is not known. In this sense, the current study explored the influence of normotensive and hypertensive postnatal environments on the intestinal microbiota structure, composition, and functional capacity of SHR and Wistar rats. Our findings revealed significant differences in the microbiota's composition and its metabolic activity in young non-fostered SHR (SS) vs. Wistar (WW) rats, even before hypertension onset, characterized by a reduction of the "low-abundance" bacterial genera, a diminished availability of fecal butyrate and elevated hydrogen sulfide (H2S) production by the SS gut microbiota. Despite influencing the microbiota of both strains, cross-fostering did not fully replicate the microbiota composition of the naturally reared groups in the SHR nursed by Wistar mothers (SW), or in the Wistar rats breastfed by SHR mothers (WS). The SW group had fewer significant genera identified at the Partial Least Squares Discriminant Analysis (PLS-DA), despite resembling the genera profile identified in the normotensive group. While sharing bacterial genera with both SS and WW groups, the WS group is distinguished by its unique microbial composition, particularly by a greater diversity of the 'low-abundance' bacterial genera. Moreover, decreased systolic blood pressure was observed in the SW group compared to the SS group in adulthood. Thus, we could establish a link between microbiota composition and hypertension development, associating it with the loss of the "low-abundance" bacterial taxa. Our data suggest that the postnatal environment is pivotal to promoting gut microbiota compositional changes and contributes to hypertension development.
Collapse
Affiliation(s)
- Patrizia Dardi
- Laboratory of Vascular Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Camille Perella Coutinho
- Department of Food and Experimental Nutrition /FoRC - Food Research Center, University of Sao Paulo, Sao Paulo, Brazil
| | - Sarah de Oliveira
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Simone Aparecida Teixeira
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Eduardo Purgatto
- Department of Food and Experimental Nutrition /FoRC - Food Research Center, University of Sao Paulo, Sao Paulo, Brazil
| | - Marco Aurélio Ramirez Vinolo
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Marcelo Nicolás Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Luciana Venturini Rossoni
- Laboratory of Vascular Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
13
|
Tursi A, Turroni S, De Bastiani R, Procaccianti G, D'Amico F, Allegretta L, Antonino N, Baldi E, Casamassima C, Casella G, Ciuffi M, De Bastiani M, Lazzarotto L, Licci C, Mancuso M, Penna A, Pranzo G, Sanna G, Tosetti C, Zamparella M, Picchio M. Gut microbiota in symptomatic uncomplicated diverticular disease stratifies by severity of abdominal pain. Eur J Gastroenterol Hepatol 2025; 37:147-153. [PMID: 39514266 DOI: 10.1097/meg.0000000000002884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
OBJECTIVE Patients with symptomatic uncomplicated diverticular disease (SUDD) may have a disrupted gut microbiota. However, current data are from small sample studies, and reported associations vary widely across studies. We aimed to profile the fecal microbiota in SUDD patients enrolled in primary care. METHODS A retrospective study was conducted in SUDD ( N = 72) and asymptomatic diverticulosis (AD) ( N = 30), the latter serving as a control group. RESULTS No significant differences in alpha and beta diversity were found between SUDD and AD, but SUDD was discriminated by a higher relative abundance of the family Streptococcaceae and the genera Alistipes , Agathobacter , and Butyricimonas . Interestingly, the gut microbiota of SUDD patients stratified by the severity of abdominal pain [according to the visual analog scale (VAS)]. In particular, higher diversity and health-associated taxa (such as Bifidobacterium , Eubacterium coprostanoligenes group, and Dorea ) characterized mild (VAS score 1-3) SUDD, Proteobacteria , Veillonellaceae and Blautia moderate (VAS score 4-7) SUDD, and Prevotellaceae and Megasphaera severe (VAS score 8-10) SUDD. CONCLUSION Our analysis suggests that specific taxa may be related to SUDD, but the associations vary depending on the severity of abdominal pain. In addition to advancing our ecological understanding of this complex disease, our findings may pave the way for the incorporation of gut microbiota profiling into clinical practice to aid patient management, including stratification and treatment.
Collapse
Affiliation(s)
- Antonio Tursi
- Territorial Gastroenterology Service, Barletta-Andria-Trani Local Health Agency, Andria
- Department of Medical and Surgical Sciences, School of Medicine, Catholic University, Rome
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna
| | - Rudi De Bastiani
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | - Giorgia Procaccianti
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna
| | - Federica D'Amico
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna
| | | | - Natale Antonino
- General Pratictioner, Private Practice Gastroenterologist, Bisceglie
- Barletta-Andria-Trani Local Health Agency, Andria
| | - Elisabetta Baldi
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | - Carlo Casamassima
- Barletta-Andria-Trani Local Health Agency, Andria
- General Pratictioner, Private Practice Gastroenterologist, San Ferdinando di Puglia
| | - Giovanni Casella
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | - Mario Ciuffi
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | - Marco De Bastiani
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | | | | | - Maurizio Mancuso
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | | | - Giuseppe Pranzo
- Ambulatory for IBD Treatment, 'Valle D'Itria' Hospital, Martina Franca
| | - Guido Sanna
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | - Cesare Tosetti
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | - Maria Zamparella
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | | |
Collapse
|
14
|
Kosinski L, Engen PA, Swanson B, Villanueva M, Shaikh M, Green SJ, Naqib A, Hamaker B, Cantu-Jungles TM, Keshavarzian A. Use of a Novel Passive E-Nose to Monitor Fermentable Prebiotic Fiber Consumption. SENSORS (BASEL, SWITZERLAND) 2025; 25:797. [PMID: 39943435 PMCID: PMC11819772 DOI: 10.3390/s25030797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025]
Abstract
We developed a home-based electronic nose (E-Nose) to passively monitor volatile organic compounds (VOCs) emitted following bowel movements and assessed its validity by correlating the output with prebiotic fiber intake. Healthy, non-overweight participants followed a three-week protocol which included the following: (1) installing the E-Nose in their bathroom; (2) activating the device following each bowel movement; (3) recording their dietary intake; (4) consuming a fiber bar (RiteCarbs) containing a blend of 10 g of prebiotic fiber daily during weeks two and three; and (5) submit stool specimens at the beginning and end of the study for 16S rRNA gene sequencing and analysis. Participants' fecal microbiome displayed significantly increased relative abundance of putative total SCFA-producing genera (p = 0.0323) [total acetate-producing genera (p = 0.0214), total butyrate-producing genera (p = 0.0131)] and decreased Gram-negative proinflammatory genera (p = 0.0468). Prebiotic intervention significantly increased the participants' fiber intake (p = 0.0152), E-Nose Min/Max (p = 0.0339), and area over the curve in VOC-to-fiber output (p = 0.0044). Increased fiber intake was negatively associated (R2 = 0.53, p = 0.026) with decreased relative abundance of putative Gram-negative proinflammatory genera. This proof-of-concept study demonstrates that a prototype E-Nose can noninvasively detect a direct connection between fiber intake and VOC outputs in a home-based environment.
Collapse
Affiliation(s)
| | - Phillip A. Engen
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, USA; (P.A.E.); (B.S.); (M.V.); (M.S.); (A.N.); (A.K.)
| | - Barbara Swanson
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, USA; (P.A.E.); (B.S.); (M.V.); (M.S.); (A.N.); (A.K.)
- Rush University College of Nursing, Rush University Medical Center, Chicago, IL 60612, USA
| | - Michelle Villanueva
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, USA; (P.A.E.); (B.S.); (M.V.); (M.S.); (A.N.); (A.K.)
| | - Maliha Shaikh
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, USA; (P.A.E.); (B.S.); (M.V.); (M.S.); (A.N.); (A.K.)
| | - Stefan J. Green
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA;
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL 60612, USA
| | - Ankur Naqib
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, USA; (P.A.E.); (B.S.); (M.V.); (M.S.); (A.N.); (A.K.)
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL 60612, USA
| | - Bruce Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (B.H.); (T.M.C.-J.)
| | - Thaisa M. Cantu-Jungles
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (B.H.); (T.M.C.-J.)
| | - Ali Keshavarzian
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, USA; (P.A.E.); (B.S.); (M.V.); (M.S.); (A.N.); (A.K.)
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA;
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Physiology, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
15
|
Skoulakis A, Skoufos G, Ovsepian A, Hatzigeorgiou AG. Machine learning models reveal microbial signatures in healthy human tissues, challenging the sterility of human organs. Front Microbiol 2025; 15:1512304. [PMID: 39931275 PMCID: PMC11808598 DOI: 10.3389/fmicb.2024.1512304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/16/2024] [Indexed: 02/13/2025] Open
Abstract
Background The presence of microbes within healthy human internal organs still remains under question. Our study endeavors to discern microbial signatures within normal human internal tissues using data from the Genotype-Tissue Expression (GTEx) consortium. Machine learning (ML) models were developed to classify each tissue type based solely on microbial profiles, with the identification of tissue-specific microbial signatures suggesting the presence of distinct microbial communities inside tissues. Methods We analyzed 13,871 normal RNA-seq samples from 28 tissues obtained from the GTEx consortium. Unaligned sequencing reads with the human genome were processed using AGAMEMNON, an algorithm for metagenomic microbial quantification, with a reference database comprising bacterial, archaeal, and viral genomes, alongside fungal transcriptomes. Gradient-boosting ML models were trained to classify each tissue against all others based on its microbial profile. To validate the findings, we analyzed 38 healthy living tissue samples (samples from healthy tissues obtained from living individuals, not deceased) from an independent study, as the GTEx samples were derived from post-mortem biopsies. Results Tissue-specific microbial signatures were identified in 11 out of the 28 tissues while the signatures for 8 tissues (Muscle, Heart, Stomach, Colon tissue, Testis, Blood, Liver, and Bladder tissue) demonstrated resilience to in silico contamination. The models for Heart, Colon tissue, and Liver displayed high discriminatory performance also in the living dataset, suggesting the presence of a tissue-specific microbiome for these tissues even in a living state. Notably, the most crucial features were the fungus Sporisorium graminicola for the heart, the gram-positive bacterium Flavonifractor plautii for the colon tissue, and the gram-negative bacterium Bartonella machadoae for the liver. Conclusion The presence of tissue-specific microbial signatures in certain tissues suggests that these organs are not devoid of microorganisms even in healthy conditions and probably they harbor low-biomass microbial communities unique to each tissue. The discoveries presented here confront the enduring dogma positing the sterility of internal tissues, yet further validation through controlled laboratory experiments is imperative to substantiate this hypothesis. Exploring the microbiome of internal tissues holds promise for elucidating the pathophysiology underlying both health and a spectrum of diseases, including sepsis, inflammation, and cancer.
Collapse
Affiliation(s)
- Anargyros Skoulakis
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- Hellenic Pasteur Institute, Athens, Greece
| | - Giorgos Skoufos
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- Hellenic Pasteur Institute, Athens, Greece
| | - Armen Ovsepian
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- Hellenic Pasteur Institute, Athens, Greece
| | - Artemis G. Hatzigeorgiou
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- Hellenic Pasteur Institute, Athens, Greece
| |
Collapse
|
16
|
Iqbal NT, Khan H, Khalid A, Mahmood SF, Nasir N, Khanum I, de Siqueira I, Van Voorhis W. Chronic inflammation in post-acute sequelae of COVID-19 modulates gut microbiome: a review of literature on COVID-19 sequelae and gut dysbiosis. Mol Med 2025; 31:22. [PMID: 39849406 PMCID: PMC11756069 DOI: 10.1186/s10020-024-00986-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/01/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Long COVID or Post-acute sequelae of COVID-19 is an emerging syndrome, recognized in COVID-19 patients who suffer from mild to severe illness and do not recover completely. Most studies define Long COVID, through symptoms like fatigue, brain fog, joint pain, and headache prevailing four or more weeks post-initial infection. Global variations in Long COVID presentation and symptoms make it challenging to standardize features of Long COVID. Long COVID appears to be accompanied by an auto-immune multi-faceted syndrome where the virus or viral antigen persistence causes continuous stimulation of the immune response, resulting in multi-organ immune dysregulation. MAIN TEXT This review is focused on understanding the risk factors of Long COVID with a special emphasis on the dysregulation of the gut-brain axis. Two proposed mechanisms are discussed here. The first mechanism is related to the dysfunction of angiotensin-converting enzyme 2 receptor due to Severe Acute Respiratory Syndrome Corona Virus 2 infection, leading to impaired mTOR pathway activation, reduced AMP secretion, and causing dysbiotic changes in the gut. Secondly, gut-brain axis dysregulation accompanied by decreased production of short-chain fatty acids, impaired enteroendocrine cell function, and increased leakiness of the gut, which favors translocation of pathogens or lipopolysaccharide in circulation causing the release of pro-inflammatory cytokines. The altered Hypothalamic-Pituitary-Adrenal axis is accompanied by the reduced level of neurotransmitter, and decreased stimulation of the vagus nerve, which may cause neuroinflammation and dysregulation of serum cortisol levels. The dysbiotic microbiome in Long COVID patients is characterized by a decrease in beneficial short chain fatty acid-producing bacteria (Faecalibacterium, Ruminococcus, Dorea, and Bifidobacterium) and an increase in opportunistic bacteria (Corynebacterium, Streptococcus, Enterococcus). This dysbiosis is transient and may be impacted by interventions including probiotics, and dietary supplements. CONCLUSIONS Further studies are required to understand the geographic variation, racial and ethnic differences in phenotypes of Long COVID, the influence of viral strains on existing and emerging phenotypes, to explore long-term effects of gut dysbiosis, and gut-brain axis dysregulation, as well as the potential role of diet and probiotics in alleviating those symptoms.
Collapse
Affiliation(s)
- Najeeha Talat Iqbal
- Department of Biological and Biomedical Sciences, Department of Pediatrics and Child Health, Aga Khan University, Stadium Road, P. O Box 3500, Karachi, 74800, Pakistan.
- Department of Pediatrics & Child Health, Aga Khan University, Karachi, Pakistan.
| | - Hana Khan
- Undergraduate Medical Education (UGME), Year II, Aga Khan University, Karachi, Pakistan
| | - Aqsa Khalid
- Department of Pediatrics & Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Nosheen Nasir
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Iffat Khanum
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | | | - Wes Van Voorhis
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, USA
| |
Collapse
|
17
|
Saito Y, Sagae T. High leafy and root vegetables and high rice dietary patterns were associated with primary and secondary bile acid levels in the feces. Sci Rep 2025; 15:2092. [PMID: 39814946 PMCID: PMC11736012 DOI: 10.1038/s41598-025-86273-8] [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: 07/01/2024] [Accepted: 01/09/2025] [Indexed: 01/18/2025] Open
Abstract
Colorectal cancer has the second highest mortality among cancer sites worldwide, with increasing morbidity, high recurrence rates, and even poorer postoperative quality of life. Therefore, preventive strategies for colorectal cancer should be established. This study aimed to cross-sectionally explore dietary patterns affecting the intestinal metabolism of bile acids (BAs), a risk factor for colorectal cancer, in young Japanese women. We collected fecal samples for intestinal microbiota and BA analysis. We used the Bristol scale to determine 1-week defecation status. Moreover, the brief-type self-administered diet history questionnaire was used for habitual dietary intake status. Reduced-rank regression analysis revealed dietary patterns related to fecal BA levels. The relationship between dietary patterns and fecal BA levels was adjusted for defecation status and intestinal microbiota variables using analysis of covariance. Reduced-rank regression analysis generated two dietary pattern scores related to fecal BA levels. First, the score was associated with a greater intake of leafy and root vegetables, and higher values were associated with greater fecal cholic and chenodeoxycholic acid levels and lower deoxycholic and lithocholic acid levels. Second, the score was associated with greater rice intake and lower Western sweets, pork, beef, and egg intake, and higher values were associated with lower deoxycholic and lithocholic acid levels. These relationships remained after adjusting for intestinal microbiota and defecation status variables.
Collapse
Affiliation(s)
- Yosuke Saito
- Department of Clinical Nutrition, Faculty of Health and Wellness Sciences, Hiroshima International University, 5-1-1, Hirokoshingai, Kure, Hiroshima, 737-0112, Japan.
- Department of Human Life Sciences, Sakura No Seibo Junior College, 3-6 Hanazono-Cho, Fukushima-Shi, Fukushima, 960-8585, Japan.
| | - Toyoaki Sagae
- Department of Health and Nutrition, Yamagata Prefectural Yonezawa University of Nutrition Sciences, 6-15-1, Torimachi, Yonezawa, Yamagata, 992-0025, Japan
| |
Collapse
|
18
|
Armetta J, Li SS, Vaaben TH, Vazquez-Uribe R, Sommer MOA. Metagenome-guided culturomics for the targeted enrichment of gut microbes. Nat Commun 2025; 16:663. [PMID: 39809763 PMCID: PMC11733127 DOI: 10.1038/s41467-024-55668-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
The gut microbiome significantly impacts human health, yet cultivation challenges hinder its exploration. Here, we combine deep whole-metagenome sequencing with culturomics to selectively enrich for taxa and functional capabilities of interest. Using a modified commercial base medium, 50 growth modifications were evaluated, spanning antibiotics, physico-chemical conditions, and bioactive compounds. Whole-metagenome sequencing identified medium additives, like caffeine, that enhance taxa often associated with healthier subjects (e.g., Lachnospiraceae, Oscillospiraceae, Ruminococcaceae). We also explore the impact of modifications on the composition of cultured communities and establish a link between medium preference and microbial phylogeny. Leveraging these insights, we demonstrate that combinations of media modifications can further enhance the targeted enrichment of taxa and metabolic functions, such as Collinsella aerofaciens, or strains harboring biochemical pathways involved in dopamine metabolism. This streamlined, scalable approach unlocks the potential for selective enrichment, advancing microbiome research by understanding the impact of different cultivation parameters on gut microbes.
Collapse
Affiliation(s)
- Jeremy Armetta
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark Kgs., Lyngby, Denmark
- Novonesis A/S, Hørsholm, Denmark
| | - Simone S Li
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark Kgs., Lyngby, Denmark
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Centre to Impact Antimicrobial Resistance, Monash University, Melbourne, VIC, Australia
| | - Troels Holger Vaaben
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark Kgs., Lyngby, Denmark
| | - Ruben Vazquez-Uribe
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark Kgs., Lyngby, Denmark
- Center for Microbiology, VIB, Leuven, Belgium
| | - Morten O A Sommer
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark Kgs., Lyngby, Denmark.
| |
Collapse
|
19
|
Klinhom S, Kunasol C, Sriwichaiin S, Kerdphoo S, Chattipakorn N, Chattipakorn SC, Thitaram C. Characteristics of gut microbiota profiles in Asian elephants (Elephas maximus) with gastrointestinal disorders. Sci Rep 2025; 15:1327. [PMID: 39779898 PMCID: PMC11711614 DOI: 10.1038/s41598-025-85495-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
Colic and diarrhea are common gastrointestinal (GI) disorders in captive Asian elephants, which can severely impact health and lead to mortality. Gut dysbiosis, indicated by alterations in gut microbiome composition, can be observed in individuals with GI disorders. However, changes in gut microbial profiles of elephants with GI disorders have never been investigated. Thus, this study aimed to elucidate the profiles of gut microbiota in captive elephants with different GI symptoms. Fecal samples were collected from eighteen elephants in Chiang Mai, Thailand, including seven healthy individuals, seven with impaction colic, and four with diarrhea. The samples were subjected to DNA extraction and amplification targeting the V3-V4 region of 16S rRNA gene for next-generation sequencing analysis. Elephants with GI symptoms exhibited a decreased microbial stability, as characterized by a significant reduction in microbiota diversity within individual guts and notable differences in microbial community composition when compared with healthy elephants. These changes included a decrease in the relative abundance of specific bacterial taxa, in elephants with GI symptoms such as a reduction in genera Rubrobacter, Rokubacteria, UBA1819, Nitrospira, and MND1. Conversely, an increase in genera Lysinibacillus, Bacteroidetes_BD2-2, and the family Marinifilaceae was observed when, compared with the healthy group. Variations in taxa of gut microbiota among elephants with GI disorders indicated diverse microbial characteristics associated with different GI symptoms. This study suggests that exploring gut microbiota dynamics in elephant health and GI disorders can lead to a better understanding of food and water management for maintaining a healthy gut and ensuring the longevity of the elephants.
Collapse
Affiliation(s)
- Sarisa Klinhom
- Center of Elephant and Wildlife Health, Animal Hospital, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand
| | - Chanon Kunasol
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neurophysiology Unit, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sirawit Sriwichaiin
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neurophysiology Unit, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sasiwan Kerdphoo
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neurophysiology Unit, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neurophysiology Unit, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neurophysiology Unit, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Chatchote Thitaram
- Center of Elephant and Wildlife Health, Animal Hospital, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand.
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand.
- Elephant, Wildlife and Companion Animals Research Group, Chiang Mai University, Chiang Mai, 50100, Thailand.
| |
Collapse
|
20
|
Voigt RM, Engen PA, Villanueva M, Bambi SA, Green SJ, Naqib A, Raeisi S, Shaikh M, Hamaker BR, Cantu-Jungles TM, Pridgen SA, Held P, Keshavarzian A. Prebiotics as an adjunct therapy for posttraumatic stress disorder: a pilot randomized controlled trial. Front Neurosci 2025; 18:1477519. [PMID: 39840022 PMCID: PMC11747240 DOI: 10.3389/fnins.2024.1477519] [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: 08/07/2024] [Accepted: 12/19/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction Posttraumatic stress disorder (PTSD) is a debilitating disorder characterized by intrusive memories, avoidance, negative thoughts and moods, and heightened arousal. Many patients also report gastrointestinal symptoms. Cognitive behavioral therapy (CBT) is an evidence-based treatment approach for PTSD that successfully reduces symptoms. However, many patients still meet criteria for PTSD after treatment or continue to have symptoms indicating the need for new treatment strategies for PTSD. Patients with PTSD have a disrupted intestinal microbiome (i.e., dysbiosis) which can promote neuroinflammation; thus, modulation of the microbiome could be an alternative or adjunct treatment approach for PTSD. Methods The current study was a 12-week, double-blind, placebo-controlled trial seeking to understand if CBT combined with a microbiota-modifying, prebiotic fiber intervention would beneficially impact clinical outcomes in veterans with PTSD (n = 70). This proof-of-concept, pilot trial was designed to assess: (1) the relationship between severity of PTSD symptoms and microbiota composition and SCFA levels (i.e., acetate, propionate, butyrate), (2) if CBT treatment with a concomitant prebiotic fiber intervention would beneficially impact clinical outcomes in veterans with PTSD, (3) evaluate the feasibility and acceptability of a prebiotic intervention as an adjunct treatment to CBT, and (4) assess the impact of treatment on the intestinal microbiota and stool SCFA (i.e., mechanism). Results This study found that PTSD severity may be associated with reduced abundance of taxa capable of producing the SCFA propionate, and that a subset of individuals with PTSD may benefit from a microbiota-modifying prebiotic intervention. Conclusion This study suggests that targeting the intestinal microbiome through prebiotic supplementation could represent a promising avenue for enhancing treatment outcomes in some individuals with PTSD. Clinical trial registration https://clinicaltrials.gov/, identifier NCT05424146.
Collapse
Affiliation(s)
- Robin M. Voigt
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Phillip A. Engen
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Michelle Villanueva
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Simona A. Bambi
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Stefan J. Green
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL, United States
| | - Ankur Naqib
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL, United States
| | - Shohreh Raeisi
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Maliha Shaikh
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Bruce R. Hamaker
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, United States
| | - Thaisa M. Cantu-Jungles
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, United States
| | - Sarah A. Pridgen
- Department of Psychiatry and Behavioral Science, Rush University Medical Center, Chicago, IL, United States
| | - Philip Held
- Department of Psychiatry and Behavioral Science, Rush University Medical Center, Chicago, IL, United States
| | - Ali Keshavarzian
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
- Department of Physiology, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
21
|
Abdullah, Ahmad N, Xiao J, Tian W, Khan NU, Hussain M, Ahsan HM, Hamed YS, Zhong H, Guan R. Gingerols: Preparation, encapsulation, and bioactivities focusing gut microbiome modulation and attenuation of disease symptoms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156352. [PMID: 39740381 DOI: 10.1016/j.phymed.2024.156352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 10/17/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Gut dysbiosis, chronic diseases, and microbial recurrent infections concerns have driven the researchers to explore phytochemicals from medicinal and food homologous plants to modulate gut microbiota, mitigate diseases, and inhibit pathogens. Gingerols have attracted attention as therapeutic agents due to their diverse biological activities like gut microbiome regulation, gastro-protective, anti-inflammatory, anti-microbial, and anti-oxidative effects. PURPOSE This review aimed to summarize the gingerols health-promoting potential, specifically focusing on the regulation of gut microbiome, attenuation of disease symptoms, mechanisms of action, and signaling pathways involved. METHOD Research findings from experimental and clinical studies have been summarized regarding gingerols effects on the modulation of gut microbiome and its metabolites, and attenuation of disease symptoms. RESULTS Gingerols are phenolic compounds characterized by a common 3-methoxy-4-hydroxyphenyl moiety in their chemical structures, and further divided into different gingerol types, including gingerols (major), shogaols, paradols, gingerdiols, gingerdiones, and zingerones (minor). Advanced extraction techniques (e.g., ionic liquid-based-, enzyme-assisted-, microwave-assisted-, pressurized liquid-, ultrasound-assisted-, and supercritical fluid extractions) were reported as optimal alternatives to conventional methods for gingerols extraction. Research studies reported that gingerols positively modulated the composition of gut microbiome that helped to combat disease symptoms (e.g., obesity by decreasing weight gain- (Lactobacillus reuteri and Lachnospiraceae) and increasing weight loss associated-bacteria (Akkermansia, Muribaculaceae, and Alloprevotella). Gingerols intervention also ameliorated ulcerative colitis by increasing relative abundance of the beneficial bacteria (Akkermansia, Lachnospiraceae NK4A136, and Muribaculaceae_norank), and decreasing pathogenic microorganisms (Bacteroides, Parabacteroides, and Desulfovibrio). Emerging delivery systems (e.g., microcapsules, nanoparticles, nanostructured lipid carriers, nanoemulsions, and nanoliposomes) can enhance the bioavailability and therapeutic efficacy of gingerols by preserving their inherent properties and addressing challenges of stability, solubility, and absorption. CONCLUSION Gingerols are promising therapeutic agents to modulate gut microbiome (increase beneficial bacteria and inhibit pathogenic microbes), and attenuate chronic disease symptoms such as diabetes, colitis, obesity, oxidative stress, and cancer. Despite significant progress, challenges persist in transforming research findings into industrial applications, such as stability and solubility during processing and low bioavailability in the distal gut to impart desirable health benefits.
Collapse
Affiliation(s)
- Abdullah
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Naveed Ahmad
- Multan College of Food & Nutrition Sciences, Multan Medical & Dental College, Multan, Pakistan
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Wenni Tian
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Naveed Ullah Khan
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Muhammad Hussain
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hafiz Muhammad Ahsan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yahya Saud Hamed
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Rongfa Guan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
22
|
Wu Y, Zhang L, Zheng H, Huang W, Zhang X, Ji W, Ma R, Mao X, Huang Y, Liu X, Zeng J, Guo S, Cheng Z, Wang T, Wang M, Wang W, Huang J, Lu X, Li L, Li Y, Bai X, Wei Y, Li X, Wang X. Oral bacteriophage therapy effectively prevent and control of Clostridium perfringens type c infections in newborn piglets. Vet Microbiol 2025; 300:110330. [PMID: 39674030 DOI: 10.1016/j.vetmic.2024.110330] [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: 07/29/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Clostridium perfringens (C. perfringens) type C exhibits strong pathogenicity, often leading to swine dysentery, severely affecting the economic efficiency of the pig farming industry. Bacteriophages as bacterial viruses have many natural advantages and are potent candidates for controlling bacterial infections. In this study, a lytic C. perfringens phage designed as vB_CpeP_15N3 was isolated with the host C. perfringens type C CVCC1155, and its potential for therapy was determined in vitro and in vivo. Despite the narrow host range, phage vB_CpeP_15N3 exhibited a large burst size of 102 PFU/cell following a short latent period of 10 min. In addition, phage vB_CpeP_15N3 remained stable at temperatures ranging from 4 to 50°C and pH levels from 5 to 9 and had a strong antibacterial effect in vitro. Through whole-genome analysis, phage vB_CpeP_15N3 belongs to the family Guelinviridae, genus Brucesealvirus with no genes related to lysogeny and bacterial virulence or resistance. We further demonstrated that phage vB_CpeP_15N3 by oral administration for preventive purposes could significantly alleviate clinical symptoms and jejunal lesions of newborn piglets through the reduced colonization of C. perfringens type C in the jejunum and the level of CPB toxin in the content of jejunum in the newborn piglet model of CVCC1155 infection. In addition, phage vB_CpeP_15N3 by oral administration for preventive purposes could improve the diversity and abundance of the jejunum microbiota in newborn piglets. Moreover, the prevention by phage vB_CpeP_15N3 obtained more effective therapeutic results than phage and gentamicin treatments. Taken together, these findings suggested that phage vB_CpeP_15N3 is a promising alternative of antibiotics for preventing and controlling C. perfringens type C infection of newborn piglets.
Collapse
Affiliation(s)
- Yuxing Wu
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Lingyuan Zhang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Haodong Zheng
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Weiqian Huang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China; Guangdong Rongda Biology Co., LTD, Qingyuan, 511517, PR China
| | - Xiaochen Zhang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Wantong Ji
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Runwen Ma
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Xinyu Mao
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Yonghua Huang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Xiaoxin Liu
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Jialing Zeng
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Shaowen Guo
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Zixin Cheng
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Tong Wang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Mei Wang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Wenqi Wang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Jingjing Huang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Xuanyi Lu
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Lixuan Li
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Yinan Li
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Xue Bai
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Yixian Wei
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Xun Li
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Xiaoye Wang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China.
| |
Collapse
|
23
|
Lalli MK, Salo TE, Hakola L, Knip M, Virtanen SM, Vatanen T. Associations between dietary fibers and gut microbiome composition in the EDIA longitudinal infant cohort. Am J Clin Nutr 2025; 121:83-99. [PMID: 39551356 PMCID: PMC11747200 DOI: 10.1016/j.ajcnut.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND The infant gut microbiome undergoes rapid changes in the first year of life, supporting normal development and long-term health. Although diet shapes this process, the role of fibers in complementary foods on gut microbiome maturation is poorly understood. OBJECTIVES We explored how the transition from human milk to fibers in complementary foods shapes the taxonomic and functional maturation of the gut microbiome within the first year of life. METHODS We assessed the longitudinal and cross-sectional development of infant gut microbiomes (N = 68 infants) and metabolomes (N = 33 infants) using linear mixed models to uncover their associations to dietary fibers and their food sources. Fiber intakes were assessed with 3-d food records (months 3, 6, 9, and 12) relying on CODEX-compliant fiber fraction values, and questionnaires tracked the overall complementary food introduction. Bacterial species were identified and quantified via MetaPhlAn2 from metagenomic data, and metabolomic profiles were obtained using 4 LC-MS methods. RESULTS We identified 176 complementary food fiber-bacterial species associations. First plant-based fibers associated with microbiota compositions similar to breastfeeding, and further associated with aromatic amino acid-derived metabolites, including 5-hydroxyindoleacetic acid (total dietary fiber - complementary foods (g) - β = 3.50, CI: 2.48, 4.52, P = 6.53 × 10-5). Distinct fibers from different food categories showed unique associations with specific bacterial taxa. Key species, such as Faecalibacterium prausnitznii, associated with oat fibers (g/MJ, β = 2.18, confidence interval: 1.36, 2.84, P = 6.12 × 10-6), reflective of maturing microbial communities. Fiber intake during weaning associated with shifts in metabolite profiles, including immunomodulatory metabolites, with fiber effects observed in a source- and timing-dependent manner, implicated in gradual microbiome diversification. CONCLUSIONS Introducing complementary dietary fibers during the weaning period supports gut microbiome diversification and stabilization. Even minor dietary variations shows significant associations with microbial taxa and functions from the onset of weaning, highlighting the importance of infant dietary recommendations that support the gut microbiome maturation during early life. This trial was registered at clinicaltrials.gov as registration number NCT01735123.
Collapse
Affiliation(s)
- Marianne K Lalli
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tuuli Ei Salo
- Department of Public Health, Finnish Institute for Health and Welfare, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Leena Hakola
- Department of Public Health, Finnish Institute for Health and Welfare, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland; Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland
| | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Helsinki University Hospital, Helsinki, Finland; Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Suvi M Virtanen
- Department of Public Health, Finnish Institute for Health and Welfare, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland; Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland; Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Tommi Vatanen
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland; Broad Institute of MIT and Harvard, Cambridge, MA, United States; Liggins Institute, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
24
|
Ferrara KM, Gupta KR, Pi H. Bacterial Organelles in Iron Physiology. Mol Microbiol 2024; 122:914-928. [PMID: 39545931 DOI: 10.1111/mmi.15330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024]
Abstract
Bacteria were once thought to be simple organisms, lacking the membrane-bound organelles found in eukaryotic cells. However, recent advancements in microscopy have changed this view, revealing a diverse array of organelles within bacterial cells. These organelles, surrounded by lipid bilayers, protein-lipid monolayers, or proteinaceous shells, play crucial roles in facilitating biochemical reactions and protecting cells from harmful byproducts. Unlike eukaryotic organelles, which are universally present, bacterial organelles are species-specific and induced only under certain conditions. This review focuses on the bacterial organelles that contain iron, an essential micronutrient for all life forms but potentially toxic when present in excess. To date, three types of iron-related bacterial organelles have been identified: two membrane-bound organelles, magnetosomes and ferrosomes, and one protein-enclosed organelle, the encapsulated ferritin-like proteins. This article provides an updated overview of the genetics, biogenesis, and physiological functions of these organelles. Furthermore, we discuss how bacteria utilize these specialized structures to adapt, grow, and survive under various environmental conditions.
Collapse
Affiliation(s)
- Kristina M Ferrara
- Department of Microbial Pathogenesis, Microbial Sciences Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kuldeepkumar R Gupta
- Department of Microbial Pathogenesis, Microbial Sciences Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Hualiang Pi
- Department of Microbial Pathogenesis, Microbial Sciences Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
25
|
Cabezas-Cruz A, Bermúdez-Humarán LG. Exploring the relationship between Faecalibacterium duncaniae and Escherichia coli in inflammatory bowel disease (IBD): Insights and implications. Comput Struct Biotechnol J 2024; 23:1-9. [PMID: 38094217 PMCID: PMC10716368 DOI: 10.1016/j.csbj.2023.11.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 03/04/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a group of disorders characterized by an inflammation of the gastrointestinal tract (GIT) and represents a major social and economic burden. Despite ongoing research into the etiology and pathophysiology of this multifactorial disease, treatment options remain limited. From this perspective, the gut microbiota has emerged as a potential player in the pathogenesis of IBD, and animal and human studies support this hypothesis. Indeed, the human gut is one of the most complex ecological communities (composed of 1013-1014 microorganisms) that plays a critical role in human health by influencing normal physiology and disease susceptibility through its collective metabolic activities and host interactions. In addition, live probiotic bacteria present in some food products (which transit through the GIT) have been shown to interact with the host immune system and confer several health benefits. The aim of this review is to provide an overview of the link between Faecalibacterium duncaniae and Escherichia coli and IBD, highlighting the main areas of research in this field. An ecological perspective on the gut microbiota may offer new insights for the development of clinical therapies targeting this bacterial community to improve human health.
Collapse
Affiliation(s)
- Alejandro Cabezas-Cruz
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort F-94700, France
| | | |
Collapse
|
26
|
Czatzkowska M, Wolak I, Harnisz M, Korzeniewska E. Microbial diversity and biosafety judgment of digestates derived from different biogas plants for agricultural applications. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 371:123329. [PMID: 39541817 DOI: 10.1016/j.jenvman.2024.123329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/25/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
The composition of microbial communities is the key to effective anaerobic digestion (AD). The microbiome driving the AD process has been extensively researched, whereas the influence of specific substrates on the microbiome of digestate remains insufficiently investigated. Digestate has considerable potential for use in soil fertilization and bioremediation, therefore its biological safety should be monitored. Moreover, the knowledge about the composition of microbial communities and their interconnections in digestate should be extended, due to the impact on soil microbiota and its functionality. The aim of this study was a comprehensive assessment of the (1) sanitary quality, (2) core microbiome, and (3) microbial interactions in digestates collected from three full-scale agricultural biogas plants, with particular emphasis on their applicability from the perspective of the resident microbiota. Analyzed samples of digestate were derived from various substrates used for AD, including plant- and animal-based materials, and industrial waste. The study demonstrated that the phyla Bacillota, Bacteroidota, and Cloacimonadota were the most dominant in digestates regardless of the composition of the processed substrates, however, member composition at the genus level differed significantly between samples. In addition, we observed that microbial genera belonging to the less prevalent phyla play an integral role in the forming of microbial community interactions. Dominant microbial taxa with broad metabolic capabilities, potentially improving soil quality and functionality, have been identified. Moreover, we confirmed, that digestate samples were free of analyzed pathogenic bacteria and parasites. The study results indicate that digestate may have an immense fertilizing and bioremediation potential that has not been fully availed of to date.
Collapse
Affiliation(s)
- Małgorzata Czatzkowska
- Department of Water Protection Engineering and Environmental Microbiology, Faculty of Geoengineering, University of Warmia and Mazury in Olsztyn, Prawocheńskiego 1, 10-720, Olsztyn, Poland.
| | - Izabela Wolak
- Department of Water Protection Engineering and Environmental Microbiology, Faculty of Geoengineering, University of Warmia and Mazury in Olsztyn, Prawocheńskiego 1, 10-720, Olsztyn, Poland.
| | - Monika Harnisz
- Department of Water Protection Engineering and Environmental Microbiology, Faculty of Geoengineering, University of Warmia and Mazury in Olsztyn, Prawocheńskiego 1, 10-720, Olsztyn, Poland.
| | - Ewa Korzeniewska
- Department of Water Protection Engineering and Environmental Microbiology, Faculty of Geoengineering, University of Warmia and Mazury in Olsztyn, Prawocheńskiego 1, 10-720, Olsztyn, Poland.
| |
Collapse
|
27
|
Fallah A, Sedighian H, Kachuei R, Fooladi AAI. Human microbiome in post-acute COVID-19 syndrome (PACS). CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 8:100324. [PMID: 39717208 PMCID: PMC11665312 DOI: 10.1016/j.crmicr.2024.100324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
The global COVID-19 pandemic, which began in 2019, is still ongoing. SARS-CoV-2, also known as the severe acute respiratory syndrome coronavirus 2, is the causative agent. Diarrhea, nausea, and vomiting are common GI symptoms observed in a significant number of COVID-19 patients. Additionally, the respiratory and GI tracts express high level of transmembrane protease serine 2 (TMPRSS2) and angiotensin-converting enzyme-2 (ACE2), making them primary sites for human microbiota and targets for SARS-CoV-2 infection. A growing body of research indicates that individuals with COVID-19 and post-acute COVID-19 syndrome (PACS) exhibit considerable alterations in their microbiome. In various human disorders, including diabetes, obesity, cancer, ulcerative colitis, Crohn's disease, and several viral infections, the microbiota play a significant immunomodulatory role. In this review, we investigate the potential therapeutic implications of the interactions between host microbiota and COVID-19. Microbiota-derived metabolites and components serve as primary mediators of microbiota-host interactions, influencing host immunity. We discuss the various mechanisms through which these metabolites or components produced by the microbiota impact the host's immune response to SARS-CoV-2 infection. Additionally, we address confounding factors in microbiome studies. Finally, we examine and discuss about a range of potential microbiota-based prophylactic measures and treatments for COVID-19 and PACS, as well as their effects on clinical outcomes and disease severity.
Collapse
Affiliation(s)
- Arezoo Fallah
- Department of Bacteriology and Virology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Rosenberg E. Diversity of bacteria within the human gut and its contribution to the functional unity of holobionts. NPJ Biofilms Microbiomes 2024; 10:134. [PMID: 39580487 PMCID: PMC11585559 DOI: 10.1038/s41522-024-00580-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/07/2024] [Indexed: 11/25/2024] Open
Abstract
The composition of bacteria in the human colon has been a subject of interest since the beginning of microbiology. With the development of methods for culturing strict anaerobic bacteria under multiple culture conditions, it was shown the gut contained more than 400 bacterial species and different people harbor different abundant species. The term "gut microbiome" in this review refers to bacteria studied in stool samples. Molecular methods for determining the bacterial composition of human gut has revealed more than 3000 species and less than 130 genera, indicating that the diversity of human colonic bacteria is concentrated at the species and strain levels. This review concludes with a discussion of how diversity can lead to unity of individual holobionts, between holobionts, and between populations. One of the reasons for the unity is that different bacterial species can have similar functional genes.
Collapse
Affiliation(s)
- Eugene Rosenberg
- Department of Microbiology, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
29
|
Senaprom S, Namjud N, Ondee T, Bumrungpert A, Pongpirul K. Sugar Composition of Thai Desserts and Their Impact on the Gut Microbiome in Healthy Volunteers: A Randomized Controlled Trial. Nutrients 2024; 16:3933. [PMID: 39599719 PMCID: PMC11597037 DOI: 10.3390/nu16223933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/03/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The relationship between consuming Thai desserts-predominantly composed of carbohydrates-and gut microbiome profiles remains unclear. This study aimed to evaluate the effects of consuming various Thai desserts with different GI values on the gut microbiomes of healthy volunteers. METHODS This open-label, parallel randomized clinical trial involved 30 healthy individuals aged 18 to 45 years. Participants were randomly assigned to one of three groups: Phetchaburi's Custard Cake (192 g, low-GI group, n = 10), Saraburi's Curry Puff (98 g, medium-GI group, n = 10), and Lampang's Crispy Rice Cracker (68 g, high-GI group, n = 10), each consumed alongside their standard breakfast. Fecal samples were collected at baseline and 24 h post-intervention for metagenomic analysis of gut microbiome profiles using 16S rRNA gene sequencing. RESULTS After 24 h, distinct trends in the relative abundance of various gut microbiota were observed among the dessert groups. In the high-GI dessert group, the abundance of Collinsella and Bifidobacterium decreased compared to the low- and medium-GI groups, while Roseburia and Ruminococcus showed slight increases. Correlation analysis revealed a significant negative relationship between sugar intake and Lactobacillus abundance in the medium- and high-GI groups, but not in the low-GI group. Additionally, a moderately negative association was observed between Akkermansia abundance and sugar intake in the high-GI group. These bacteria are implicated in energy metabolism and insulin regulation. LEfSe analysis identified Porphyromonadaceae and Porphyromonas as core microbiota in the low-GI group, whereas Klebsiella was enriched in the high-GI group, with no predominant bacteria identified in the medium-GI group. CONCLUSIONS The findings suggest that Thai desserts with varying GI levels can influence specific gut bacteria, though these effects may be temporary.
Collapse
Affiliation(s)
- Sayamon Senaprom
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.); (N.N.); (T.O.)
| | - Nuttaphat Namjud
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.); (N.N.); (T.O.)
| | - Thunnicha Ondee
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.); (N.N.); (T.O.)
- Center of Excellence in Preventive and Integrative Medicine (CE-PIM), Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Akkarach Bumrungpert
- College of Integrative Medicine, Dhurakij Pundit University, Bangkok 10210, Thailand;
| | - Krit Pongpirul
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.); (N.N.); (T.O.)
- Center of Excellence in Preventive and Integrative Medicine (CE-PIM), Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Bumrungrad International Hospital, Bangkok 10110, Thailand
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Infection Biology & Microbiomes, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3GB, UK
| |
Collapse
|
30
|
Wei L, Hui Y, Jinxi W, Shihui L, Hongping L, Jian L, Lin L. Zuogui Jiangtang Jieyu prescription improves diabetes-related depression by modulation of gut microbiota and neuroinflammation in hippocampus. Heliyon 2024; 10:e39291. [PMID: 39524747 PMCID: PMC11543871 DOI: 10.1016/j.heliyon.2024.e39291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Context There is a significant challenge associated with the co-morbidity of mental and physical illnesses throughout the world. A classic example of mental/physical co-morbidity is diabetes-related depression (DD). Objective DD is treated with Zuogui Jiangtang Jieyu prescription (ZJJ). Diabetes and psychiatric disorders are associated with dysbiosis of the gut microbiota. In this study, the aim is to examine the effects of ZJJ on gut microbiota and neuroinflammation in DD. Methods A model of DD was established and treated with medium and high doses of ZJJ as well as Metformin & Fluoxetine. A detection of depressive-like behavior was then conducted on the rats. Proinflammatory cytokines were measured in cerebrospinal fluid, and HPA axis-related proteins, glucose metabolism, and lipopolysaccharide (LPS) were measured in serum. Fecal samples from each group were collected and analyzed by 16S rRNA sequencing; TLR4 and MyD88 levels were detected by Western blot and immunohistochemistry (IHC) in the hippocampus. Results High doses of ZJJ (ZJJ-H) were found to alleviate HPA axis hyperactivity and improve gut microbiota in rats with DD. Additionally, ZJJ treatment attenuated the inflammatory response in cerebrospinal fluid, e.g. a significant reduction in proinflammatory factors, a decrease in serum LPS levels, and an inhibition of TLR4/MyD88-related pathways in the hippocampus. Discussion and conclusion ZJJ improved DD glucose metabolism and alleviated depression-like behaviors by improving gut microbiota and inhibiting hippocampal TLR4/Myd88 signaling pathways.
Collapse
Affiliation(s)
- Li Wei
- Medical Innovation Experimental center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yang Hui
- Medical Innovation Experimental center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wang Jinxi
- Medical Innovation Experimental center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lei Shihui
- Medical Innovation Experimental center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Long Hongping
- Medical Innovation Experimental center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liu Jian
- Medical Innovation Experimental center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liu Lin
- Medical Innovation Experimental center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
31
|
Hillege LE, Stevens MAM, Kristen PAJ, de Vos-Geelen J, Penders J, Redinbo MR, Smidt ML. The role of gut microbial β-glucuronidases in carcinogenesis and cancer treatment: a scoping review. J Cancer Res Clin Oncol 2024; 150:495. [PMID: 39537966 PMCID: PMC11561038 DOI: 10.1007/s00432-024-06028-2] [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: 09/10/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION The human gut microbiota influence critical functions including the metabolism of nutrients, xenobiotics, and drugs. Gut microbial β-glucuronidases (GUS) enzymes facilitate the removal of glucuronic acid from various compounds, potentially affecting anti-cancer drug efficacy and reactivating carcinogens. This review aims to comprehensively analyze and summarize studies on the role of gut microbial GUS in cancer and its interaction with anti-cancer treatments. Its goal is to collate and present insights that are directly relevant to patient care and treatment strategies in oncology. METHODS This scoping review followed PRISMA-ScR guidelines and focused on primary research exploring the role of GUS within the gut microbiota related to cancer etiology and anti-cancer treatment. Comprehensive literature searches were conducted in PubMed, Embase, and Web of Science. RESULTS GUS activity was only investigated in colorectal cancer (CRC), revealing increased fecal GUS activity, variations in the gut microbial composition, and GUS-contributing bacterial taxa in CRC patients versus controls. Irinotecan affects gastrointestinal (GI) health by increasing GUS expression and shifting gut microbial composition, particularly by enhancing the presence of GUS-producing bacteria, correlating with irinotecan-induced GI toxicities. GUS inhibitors (GUSi) can mitigate irinotecan's adverse effects, protecting the intestinal barrier and reducing diarrhea. CONCLUSION To our knowledge, this is the first review to comprehensively analyze and summarize studies on the critical role of gut microbial GUS in cancer and anti-cancer treatment, particularly irinotecan. It underscores the potential of GUSi to reduce side effects and enhance treatment efficacy, highlighting the urgent need for further research to integrate GUS targeting into future anti-cancer treatment strategies.
Collapse
Affiliation(s)
- Lars E Hillege
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands.
- Department of Surgery, FHML, Maastricht University Medical Center+, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands.
| | - Milou A M Stevens
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
- Department of Surgery, FHML, Maastricht University Medical Center+, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
| | - Paulien A J Kristen
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
- Department of Surgery, FHML, Maastricht University Medical Center+, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
| | - Judith de Vos-Geelen
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
- Division of Medical Oncology, Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - John Penders
- NUTRIM - School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Matthew R Redinbo
- Departments of Chemistry, Biochemistry & Biophysics, and Microbiology & Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Marjolein L Smidt
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
- Department of Surgery, FHML, Maastricht University Medical Center+, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
| |
Collapse
|
32
|
Romero-Rodríguez A, Ruíz-Villafán B, Sánchez S, Paredes-Sabja D. Is there a role for intestinal sporobiota in the antimicrobial resistance crisis? Microbiol Res 2024; 288:127870. [PMID: 39173554 DOI: 10.1016/j.micres.2024.127870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 08/24/2024]
Abstract
Antimicrobial resistance (AMR) is a complex issue requiring specific, multi-sectoral measures to slow its spread. When people are exposed to antimicrobial agents, it can cause resistant bacteria to increase. This means that the use, misuse, and excessive use of antimicrobial agents exert selective pressure on bacteria, which can lead to the development of "silent" reservoirs of antimicrobial resistance genes. These genes can later be mobilized into pathogenic bacteria and contribute to the spread of AMR. Many socioeconomic and environmental factors influence the transmission and dissemination of resistance genes, such as the quality of healthcare systems, water sanitation, hygiene infrastructure, and pollution. The sporobiota is an essential part of the gut microbiota that plays a role in maintaining gut homeostasis. However, because spores are highly transmissible and can spread easily, they can be a vector for AMR. The sporobiota resistome, particularly the mobile resistome, is important for tracking, managing, and limiting the spread of antimicrobial resistance genes among pathogenic and commensal bacterial species.
Collapse
Affiliation(s)
- A Romero-Rodríguez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Ciudad de México 04510, Mexico.
| | - B Ruíz-Villafán
- Laboratorio de Microbiología Industrial. Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - S Sánchez
- Laboratorio de Microbiología Industrial. Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - D Paredes-Sabja
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
33
|
Kou R, Mi F, Peng C, Ding X, Meng C, Liu F, Xiong L. Structural characterization and immunomodulatory activity of polysaccharides from the lateral roots of Aconitum carmichaelii. Int J Biol Macromol 2024; 282:136935. [PMID: 39490860 DOI: 10.1016/j.ijbiomac.2024.136935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/27/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Two polysaccharides, named FPS1-1 and FPS1-2, were separated from the neutral polysaccharides of the lateral roots of Aconitum carmichaelii, a widely used traditional Chinese medicine (Fuzi in Chinese). The monosaccharide composition analysis indicated that both FPS1-1 and FPS1-2 were glucans. However, further physicochemical analysis of FPS1-1 and FPS1-2 revealed distinct properties between the two glucans. FPS1-1 had a molecular weight (Mw) of 106.23 kDa with a spherical conformation, while FPS1-2 had a lower Mw of 19.23 kDa with a random coil conformation. The structure of FPS1-2 was further determined as a glucan whose backbone structure was composed of →4)-α-D-Glcp-(1→. The immunological activities of two polysaccharides were evaluated by a cyclophosphamide (CTX)-induced immunodeficiency model in mice. The result showed that FPS1-2 could restore CTX-induced immunosuppression by modulating CD4+ T cells differentiation and promoting cytokine secretion. Notably, FPS1-2 could modulate the colonic short-chain fatty acid (SCFA) levels and reverse the gut microbial dysbiosis induced by CTX. These findings reveal the potential benefits of Fuzi polysaccharides and provide evidences for developing immunologically functional products from Fuzi polysaccharides.
Collapse
Affiliation(s)
- Renbo Kou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fuxin Mi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xingjie Ding
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chunwang Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fei Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Liang Xiong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
34
|
Chaudhary S, Kaur P, Singh TA, Bano KS, Vyas A, Mishra AK, Singh P, Mehdi MM. The dynamic crosslinking between gut microbiota and inflammation during aging: reviewing the nutritional and hormetic approaches against dysbiosis and inflammaging. Biogerontology 2024; 26:1. [PMID: 39441393 DOI: 10.1007/s10522-024-10146-2] [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: 08/02/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024]
Abstract
The early-life gut microbiota (GM) is increasingly recognized for its contributions to human health and disease over time. Microbiota composition, influenced by factors like race, geography, lifestyle, and individual differences, is subject to change. The GM serves dual roles, defending against pathogens and shaping the host immune system. Disruptions in microbial composition can lead to immune dysregulation, impacting defense mechanisms. Additionally, GM aids digestion, releasing nutrients and influencing physiological systems like the liver, brain, and endocrine system through microbial metabolites. Dysbiosis disrupts intestinal homeostasis, contributing to age-related diseases. Recent studies are elucidating the bacterial species that characterize a healthy microbiota, defining what constitutes a 'healthy' colonic microbiota. The present review article focuses on the importance of microbiome composition for the development of homeostasis and the roles of GM during aging and the age-related diseases caused by the alteration in gut microbial communities. This article might also help the readers to find treatments targeting GM for the prevention of various diseases linked to it effectively.
Collapse
Affiliation(s)
- Sakshi Chaudhary
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Pardeep Kaur
- Department of Biotechnology, Chandigarh University, Mohali, Punjab, 140413, India
| | - Thokchom Arjun Singh
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Kaniz Shahar Bano
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Ashish Vyas
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Alok Kumar Mishra
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Prabhakar Singh
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India
| | - Mohammad Murtaza Mehdi
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| |
Collapse
|
35
|
Al-Kabe SH, Niamah AK. Current Trends and Technological Advancements in the Use of Oxalate-Degrading Bacteria as Starters in Fermented Foods-A Review. Life (Basel) 2024; 14:1338. [PMID: 39459637 PMCID: PMC11509417 DOI: 10.3390/life14101338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/15/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
Nephrolithiasis is a medical condition characterized by the existence or development of calculi, commonly referred to as stones within the renal system, and poses significant health challenges. Calcium phosphate and calcium oxalate are the predominant constituents of renal calculi and are introduced into the human body primarily via dietary sources. The presence of oxalates can become particularly problematic when the delicate balance of the normal flora residing within the gastrointestinal tract is disrupted. Within the human gut, species of Oxalobacter, Lactobacillus, and Bifidobacterium coexist in a symbiotic relationship. They play a pivotal role in mitigating the risk of stone formation by modulating certain biochemical pathways and producing specific enzymes that can facilitate the breakdown and degradation of oxalate salts. The probiotic potential exhibited by these bacteria is noteworthy, as it underscores their possible utility in the prevention of nephrolithiasis. Investigating the mechanisms by which these beneficial microorganisms exert their effects could lead to novel therapeutic strategies aimed at reducing the incidence of kidney stones. The implications of utilizing probiotics as a preventive measure against kidney stone formation represent an intriguing frontier in both nephrology and microbiome research, meriting further investigation to unlock their full potential.
Collapse
Affiliation(s)
| | - Alaa Kareem Niamah
- Department of Food Science, College of Agriculture, University of Basrah, Basra City 61004, Iraq;
| |
Collapse
|
36
|
Yafarova AA, Dementeva EV, Zlobovskaya OA, Sheptulina AF, Lopatukhina EV, Timofeev YS, Glazunova EV, Lyundup AV, Doludin YV, Kiselev AR, Shipulin GA, Makarov VV, Drapkina OM, Yudin SM. Gut Microbiota and Metabolic Alterations Associated with Heart Failure and Coronary Artery Disease. Int J Mol Sci 2024; 25:11295. [PMID: 39457077 PMCID: PMC11508380 DOI: 10.3390/ijms252011295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
This study investigates the role of gut microbiota in cardiovascular diseases, with an additional focus on pro-atherogenic metabolites. We use advanced network analysis and machine learning techniques to identify key microbial features linked to coronary artery disease (CAD) and heart failure with reduced ejection fraction (HFrEF). This cross-sectional study included 189 participants divided into three groups: coronary artery disease (n = 93), heart failure with reduced ejection fraction (n = 43), and controls (n = 53). Assessments included physical exams, echocardiography, dietary surveys, blood analysis, and fecal analysis. Gut microbiota composition was analyzed using next-generation sequencing (NGS) and quantitative polymerase chain reaction (qPCR). Statistical analysis methods for testing hypotheses and correlations, alpha and beta-diversity analyses, co-occurrence networks, and machine learning were conducted using Python libraries or R packages with multiple comparisons corrected using the Benjamini-Hochberg procedure. Significant gut microbiota alterations were observed, with higher Bacillota/Bacteroidota ratios in CAD and HFrEF groups compared to controls (p < 0.001). Significant differences were observed in α-diversity indices (Pielou, Chao1, Faith) between disease groups and controls (p < 0.001). β-diversity analyses also revealed distinct microbial profiles (p = 0.0015). Interestingly, trimethylamine N-oxide (TMAO) levels were lower in CAD and HFrEF groups compared to controls (p < 0.05), while indoxyl sulfate (IS) levels were comparable between the study groups. Co-occurrence network analysis and machine learning identified key microbial features linked to these conditions, highlighting complex interactions within the gut microbiota associated with cardiovascular disease.
Collapse
Affiliation(s)
- Adel A. Yafarova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Elena V. Dementeva
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Olga A. Zlobovskaya
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Anna F. Sheptulina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Elena V. Lopatukhina
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Yuriy S. Timofeev
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Evgeniya V. Glazunova
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Aleksey V. Lyundup
- Endocrinology Research Centre, Dmitry Ulyanov St. 19, 117036 Moscow, Russia
| | - Yuriy V. Doludin
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Anton R. Kiselev
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - German A. Shipulin
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Valentin V. Makarov
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Oxana M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Sergey M. Yudin
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| |
Collapse
|
37
|
Dou B, Wu X, He Y, Xu G, Zhang H, Huang Q, Chen X, Duan N, Zhou L, Zhang W, An H, Zheng Y. Fei-Yan-Qing-Hua decoction attenuates influenza virus infection by enhancing host antiviral response through microbiota-derived acetate. Front Pharmacol 2024; 15:1446749. [PMID: 39449967 PMCID: PMC11499185 DOI: 10.3389/fphar.2024.1446749] [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: 06/10/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Background Fei-Yan-Qing-Hua decoction (FYQHD) is derived from the well-known Ma Xing Shi Gan decoction, which was documented in Zhang Zhong Jing's "Treatise on Exogenous Febrile Disease" during the Han Dynasty. Although FYQHD has been used in the treatment of pneumonia and has demonstrated clinical efficacy for decades, the underlying mechanism by which FYQHD protects against influenza virus infection through modulation of gut flora remains unclear. Here, we examined the regulatory impacts of FYQHD on an influenza virus-infected mouse model and explored the mechanisms involved. Methods An infectious mouse model was created by intranasal instillation of influenza A virus (IAV). The effectiveness of FYQHD was assessed through various measures, including weight loss, lung wet/dry ratio, oxidative stress levels, viral load in lung tissues, and intestinal injuries. Changes in gut microbiota and SCFA production were also examined. Results The results showed that FYQHD significantly reduced viral load, increased the production of type I interferon (IFN-I), and restored the integrity of the intestinal barrier following IAV challenge. Additionally, FYQHD significantly corrected the dysbiosis of gut microbiota induced by influenza virus infection, enhancing the abundance of SCFA-producing bacteria and acetate production. However, the depletion of gut microbiota significantly attenuated the protective effects of FYQHD against influenza virus infection. In vitro, the antiviral effect of acetate was demonstrated through the upregulation of concentrations of IFN-β. Conclusion FYQHD attenuates influenza virus-induced lung and intestinal injuries by boosting the host antiviral response through increasing the abundance of Lachnospiraceae_NK4A136 and Roseburia, along with elevated acetate levels. The study advances our understanding of the therapeutic mechanisms of FYQHD and provides a theoretical basis for the application of FYQHD in the treatment of influenza.
Collapse
Affiliation(s)
- Biao Dou
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Medical Laboratory, The First Affiliated Hospital of Henan University, Kaifeng, China
| | - Xiao Wu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yurong He
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guihua Xu
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huan Zhang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qilin Huang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuan Chen
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Naifan Duan
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Linqiong Zhou
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Zhang
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huazhang An
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
38
|
Salehi S, Allahverdy J, Pourjafar H, Sarabandi K, Jafari SM. Gut Microbiota and Polycystic Ovary Syndrome (PCOS): Understanding the Pathogenesis and the Role of Probiotics as a Therapeutic Strategy. Probiotics Antimicrob Proteins 2024; 16:1553-1565. [PMID: 38421576 DOI: 10.1007/s12602-024-10223-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 03/02/2024]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common disorders among women in modern societies. A variety of factors can contribute to the development of PCOS. These women often exhibit high insulin resistance (IR), hyperandrogenism, irregular periods, and infertility. Dysbiosis of the gut microbiota (GMB) in women with PCOS has attracted the attention of many researchers. Porphyromonas spp., B. coprophilus, and F. prausnitzii are found in higher numbers in the gut of women with PCOS. Short-chain fatty acids (SCFAs), produced by the intestinal microbiota through fermentation, play an essential role in regulating metabolic activities and are helpful in reducing insulin resistance and improving PCOS symptoms. According to studies, the bacteria producing SCFAs in the gut of these women are less abundant than in healthy women. The effectiveness of using probiotic supplements has been proven to improve the condition of women with PCOS. Daily consumption of probiotics improves dysbiosis of the intestinal microbiome and increases the production of SCFAs.
Collapse
Affiliation(s)
- Samaneh Salehi
- Department of Food Biotechnology, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Javad Allahverdy
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Students' Research Committee, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hadi Pourjafar
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Khashayar Sarabandi
- Research Institute of Food Science and Technology (RIFST), Km 12 Mashhad-Quchan Highway, PO Box 91895, Mashhad, 157-356, Iran
| | - Seid Mahdi Jafari
- Department of Food Materials and Process Design Engineering, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran.
- Halal Research Center of IRI, Iran Food and Drug Administration, Ministry of Health and Medical Education, Tehran, Iran.
| |
Collapse
|
39
|
Lages da Silva DH, Marques da Silva RL, Rios DL, de Souza DDG, Aburjaile F, de Freitas Neto OC, Camargos Lara LJ, Dias Araújo M, Ecco R. Intestinal microbiota diversity from broilers with runting and stunting syndrome performed by metagenomics. Avian Pathol 2024; 53:408-418. [PMID: 38662518 DOI: 10.1080/03079457.2024.2348509] [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: 11/29/2023] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 06/05/2024]
Abstract
Runting and stunting syndrome (RSS) is an enteric viral disease in commercial poultry that directly affects gut health; however, its influence on gut microbiota remains unknown. This study aimed to investigate the compositional changes in the bacterial community of the ileum of 7-day-old broiler chicks naturally affected or not affected by RSS, using next-generation sequencing (NGS) technology. Twenty-one samples were obtained from the ileal contents and mucosa of 11 chicks with RSS and 10 healthy chicks, raised in a dark house system located on a farm in the state of Minas Gerais, Brazil. The results revealed overall changes in the gut microbiota of the chicks with RSS, including a decrease in microbial richness and diversity. In particular, there was a decrease in Lactobacillus and an increase in Candidatus Arthromitus and Clostridium sensu stricto 1. These results indicate a relationship between viral infection and the gut microbial composition, which can cause gut dysbiosis and may influence inflammation in this organ.RESEARCH HIGHLIGHTS RSS causes dysbiosis of the gut microbiota of the ilea of chicks.A difference was found in gut microbiota between chicks with or without RSS.Candidatus Arthromitus was predominant in chicks with RSS.Clostridium sensu stricto 1 was strictly associated with chicks with RSS.
Collapse
Affiliation(s)
- Dayse Helena Lages da Silva
- Pathology Sector and MULTILAB, Department of Veterinary Clinic and Surgery, Veterinary School, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Rodrigo Luiz Marques da Silva
- Pathology Sector and MULTILAB, Department of Veterinary Clinic and Surgery, Veterinary School, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Diego Lisboa Rios
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Danielle da Glória de Souza
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Flávia Aburjaile
- Department of Preventive Veterinary Medicine, Veterinary School, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Oliveiro Caetano de Freitas Neto
- Department of Preventive Veterinary Medicine, Veterinary School, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Leonardo José Camargos Lara
- Department of Zootechnics, Veterinary School, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Matheus Dias Araújo
- Pathology Sector and MULTILAB, Department of Veterinary Clinic and Surgery, Veterinary School, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Roselene Ecco
- Pathology Sector and MULTILAB, Department of Veterinary Clinic and Surgery, Veterinary School, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
40
|
Ahmed I, Zhuang Z, Umar Farooq M, Li H, Wang S, Zhong Y, Zhang L, Zhang B. Efficient reduction of antibiotic resistance genes and mobile genetic elements in organic waste composting via fenton-like treatment. BIORESOURCE TECHNOLOGY 2024; 410:131306. [PMID: 39155020 DOI: 10.1016/j.biortech.2024.131306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Livestock manure harbors antibiotic resistance genes (ARGs), and aerobic composting (AC) is widely adopted for waste management. However, mitigating ARG resurgence in later stages remains challenging. This work aims to curb ARGs rebounding through a Fenton-like reaction during food waste and swine manure co-composting. Results revealed that 0.025 % zerovalent iron (ZVI) + 0.5 % hydrogen peroxide (H2O2) facilitated maximum ARG, mobile genetic elements (MGEs), and 16 s rRNA removal with reductions of 2.68, 2.69, and 1.4 logs. Spectroscopic analysis confirmed Fenton-like reaction and cell apoptosis analysis indicated that 0.025 % ZVI and 0.5 % H2O2 treatment had the maximum early apoptosis, least observed, and normal cells on day 30. Redundancy analysis highlighted the influence of bacterial communities and physicochemical properties on ARGs, with MGEs playing a crucial role in Fenton treatments. Our findings suggest incorporating ZVI and H2O2 in composting can significantly reduce ARGs and enhance waste management practices.
Collapse
Affiliation(s)
- Imtiaz Ahmed
- School of Environmental Science and Engineering, Shanghai Jiaotong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Zixian Zhuang
- School of Environmental Science and Engineering, Shanghai Jiaotong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Muhammad Umar Farooq
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Hu Li
- School of Ecology and Environment, Ningxia University, Yinchuan 750021, China
| | - Shiquan Wang
- School of Ecology and Environment, Ningxia University, Yinchuan 750021, China
| | - Yanxia Zhong
- School of Ecology and Environment, Ningxia University, Yinchuan 750021, China
| | - Lizhi Zhang
- School of Environmental Science and Engineering, Shanghai Jiaotong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Bo Zhang
- School of Environmental Science and Engineering, Shanghai Jiaotong University, 800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
41
|
Zhang C, Ma M, Zhao Z, Feng Z, Chu T, Wang Y, Liu J, Wan X. Gut mucosal microbiota profiles linked to development of positional-specific human colorectal cancer. AIMS Microbiol 2024; 10:812-832. [PMID: 39628718 PMCID: PMC11609426 DOI: 10.3934/microbiol.2024035] [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: 03/19/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 12/06/2024] Open
Abstract
Colorectal cancer (CRC) continuously ranks as the third most common cause of cancer-related deaths worldwide. Based on anatomical classifications and clinical diagnoses, CRC is classified into right-sided, left-sided, and rectal CRC. Importantly, the three types of positional-specific CRC affect the prognosis outcomes, thus indicating that positional-specific treatments for CRC are required. Emerging evidence suggests that besides host genetic and epigenetic alterations, gut mucosal microbiota is linked to gut inflammation, CRC occurrence, and prognoses. However, gut mucosal microbiota associated with positional-specific CRC are poorly investigated. Here, we report the gut mucosal microbiota profiles associated with these three types of CRC. Our analysis showed that the unique composition and biodiversity of bacterial taxa are linked to positional-specific CRC. We found that a combination of bacterial taxa can serve as potential biomarkers to distinguish the three types of CRC. Further investigations of the physiological roles of bacteria associated with positional-specific CRC may help understand the mechanism of CRC progression in different anatomical locations under the impact of gut mucosal microbiota.
Collapse
Affiliation(s)
- Chunze Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, China
- Tianjin Institute of Coloproctology, Tianjin, China
| | - Mingqian Ma
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhenying Zhao
- Department of Colorectal Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Zhiqiang Feng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Tianhao Chu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yijia Wang
- Tianjin institute of spinal surgery, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Jun Liu
- Department of Radiology, The Fourth Central Hospital Affiliated to Nankai University, Tianjin, China
| | - Xuehua Wan
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, China
| |
Collapse
|
42
|
Deyaert S, Poppe J, Dai Vu L, Baudot A, Bubeck S, Bayne T, Krishnan K, Giusto M, Moltz S, Van den Abbeele P. Functional Muffins Exert Bifidogenic Effects along with Highly Product-Specific Effects on the Human Gut Microbiota Ex Vivo. Metabolites 2024; 14:497. [PMID: 39330504 PMCID: PMC11433953 DOI: 10.3390/metabo14090497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024] Open
Abstract
GoodBiome™ Foods are functional foods containing a probiotic (Bacillus subtilis HU58™) and prebiotics (mainly inulin). Their effects on the human gut microbiota were assessed using ex vivo SIFR® technology, which has been validated to provide clinically predictive insights. GoodBiome™ Foods (BBM/LCM/OSM) were subjected to oral, gastric, and small intestinal digestion/absorption, after which their impact on the gut microbiome of four adults was assessed (n = 3). All GoodBiome™ Foods boosted health-related SCFA acetate (+13.1/14.1/13.8 mM for BBM/LCM/OSM), propionate (particularly OSM; +7.4/7.5/8.9 mM for BBM/LCM/OSM) and butyrate (particularly BBM; +2.6/2.1/1.4 mM for BBM/LCM/OSM). This is related to the increase in Bifidobacterium species (B. catenulatum, B. adolescentis, B. pseudocatenulatum), Coprococcus catus and Bacteroidetes members (Bacteroides caccae, Phocaeicola dorei, P. massiliensis), likely mediated via inulin. Further, the potent propionogenic potential of OSM related to increased Bacteroidetes members known to ferment oats (s key ingredient of OSM), while the butyrogenic potential of BBM related to a specific increase in Anaerobutyricum hallii, a butyrate producer specialized in the fermentation of erythritol (key ingredient of BBM). In addition, OSM/BBM suppressed the pathogen Clostridioides difficile, potentially due to inclusion of HU58™ in GoodBiome™ Foods. Finally, all products enhanced a spectrum of metabolites well beyond SCFA, including vitamins (B3/B6), essential amino acids, and health-related metabolites such as indole-3-propionic acid. Overall, the addition of specific ingredients to complex foods was shown to specifically modulate the gut microbiome, potentially contributing to health benefits. Noticeably, our findings contradict a recent in vitro study, underscoring the critical role of employing a physiologically relevant digestion/absorption procedure for a more accurate evaluation of the microbiome-modulating potential of complex foods.
Collapse
Affiliation(s)
- Stef Deyaert
- Cryptobiotix, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium; (S.D.)
| | - Jonas Poppe
- Cryptobiotix, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium; (S.D.)
| | - Lam Dai Vu
- Cryptobiotix, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium; (S.D.)
| | - Aurélien Baudot
- Cryptobiotix, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium; (S.D.)
| | - Sarah Bubeck
- Bubeck Scientific Communications, 194 Rainbow Drive #9418, Livingston, TX 77399, USA
| | - Thomas Bayne
- Microbiome Labs, 101 E Town Pl, Saint Augustine, FL 92092, USA
| | - Kiran Krishnan
- Microbiome Labs, 101 E Town Pl, Saint Augustine, FL 92092, USA
| | - Morgan Giusto
- Microbiome Labs, 101 E Town Pl, Saint Augustine, FL 92092, USA
| | - Samuel Moltz
- Novonesis, Biologiens Vej 2, 2800 Lyngby, Denmark
| | | |
Collapse
|
43
|
Van Hul M, Neyrinck AM, Everard A, Abot A, Bindels LB, Delzenne NM, Knauf C, Cani PD. Role of the intestinal microbiota in contributing to weight disorders and associated comorbidities. Clin Microbiol Rev 2024; 37:e0004523. [PMID: 38940505 PMCID: PMC11391702 DOI: 10.1128/cmr.00045-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024] Open
Abstract
SUMMARYThe gut microbiota is a major factor contributing to the regulation of energy homeostasis and has been linked to both excessive body weight and accumulation of fat mass (i.e., overweight, obesity) or body weight loss, weakness, muscle atrophy, and fat depletion (i.e., cachexia). These syndromes are characterized by multiple metabolic dysfunctions including abnormal regulation of food reward and intake, energy storage, and low-grade inflammation. Given the increasing worldwide prevalence of obesity, cachexia, and associated metabolic disorders, novel therapeutic strategies are needed. Among the different mechanisms explaining how the gut microbiota is capable of influencing host metabolism and energy balance, numerous studies have investigated the complex interactions existing between nutrition, gut microbes, and their metabolites. In this review, we discuss how gut microbes and different microbiota-derived metabolites regulate host metabolism. We describe the role of the gut barrier function in the onset of inflammation in this context. We explore the importance of the gut-to-brain axis in the regulation of energy homeostasis and glucose metabolism but also the key role played by the liver. Finally, we present specific key examples of how using targeted approaches such as prebiotics and probiotics might affect specific metabolites, their signaling pathways, and their interactions with the host and reflect on the challenges to move from bench to bedside.
Collapse
Affiliation(s)
- Matthias Van Hul
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, France/Belgium
| | - Audrey M Neyrinck
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
| | - Amandine Everard
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | | | - Laure B Bindels
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Nathalie M Delzenne
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
| | - Claude Knauf
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, France/Belgium
- INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), Université Paul Sabatier, Toulouse III, CHU Purpan, Toulouse, France
| | - Patrice D Cani
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, France/Belgium
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), Brussels, Belgium
| |
Collapse
|
44
|
Ling J, Hryckowian AJ. Re-framing the importance of Group B Streptococcus as a gut-resident pathobiont. Infect Immun 2024; 92:e0047823. [PMID: 38436256 PMCID: PMC11392526 DOI: 10.1128/iai.00478-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Streptococcus agalactiae (Group B Streptococcus, GBS) is a Gram-positive bacterial species that causes disease in humans across the lifespan. While antibiotics are used to mitigate GBS infections, it is evident that antibiotics disrupt human microbiomes (which can predispose people to other diseases later in life), and antibiotic resistance in GBS is on the rise. Taken together, these unintended negative impacts of antibiotics highlight the need for precision approaches for minimizing GBS disease. One possible approach involves selectively depleting GBS in its commensal niches before it can cause disease at other body sites or be transmitted to at-risk individuals. One understudied commensal niche of GBS is the adult gastrointestinal (GI) tract, which may predispose colonization at other body sites in individuals at risk for GBS disease. However, a better understanding of the host-, microbiome-, and GBS-determined variables that dictate GBS GI carriage is needed before precise GI decolonization approaches can be developed. In this review, we synthesize current knowledge of the diverse body sites occupied by GBS as a pathogen and as a commensal. We summarize key molecular factors GBS utilizes to colonize different host-associated niches to inform future efforts to study GBS in the GI tract. We also discuss other GI commensals that are pathogenic in other body sites to emphasize the broader utility of precise de-colonization approaches for mitigating infections by GBS and other bacterial pathogens. Finally, we highlight how GBS treatments could be improved with a more holistic understanding of GBS enabled by continued GI-focused study.
Collapse
Affiliation(s)
- Joie Ling
- Department of
Medicine, Division of Gastroenterology and Hepatology, University of
Wisconsin School of Medicine and Public
Health, Madison,
Wisconsin, USA
- Department of Medical
Microbiology and Immunology, University of Wisconsin School of Medicine
and Public Healthon,
Madison, Wisconsin, USA
- Microbiology Doctoral
Training Program, University of
Wisconsin-Madison, Madison,
Wisconsin, USA
| | - Andrew J. Hryckowian
- Department of
Medicine, Division of Gastroenterology and Hepatology, University of
Wisconsin School of Medicine and Public
Health, Madison,
Wisconsin, USA
- Department of Medical
Microbiology and Immunology, University of Wisconsin School of Medicine
and Public Healthon,
Madison, Wisconsin, USA
| |
Collapse
|
45
|
Passos FC, de Oliveira LMG, Jesus FR, Zanette DL, Neto OLL, Neves MCLC, Lemos ACM, Baccan GC. Beneficial Bacteria in the Gut Microbiota May Lead to Improved Metabolic and Immunological Status in Chronic Obstructive Pulmonary Disease. Med Sci (Basel) 2024; 12:41. [PMID: 39189204 PMCID: PMC11348168 DOI: 10.3390/medsci12030041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/08/2024] [Accepted: 08/09/2024] [Indexed: 08/28/2024] Open
Abstract
The progression of chronic obstructive pulmonary disease (COPD) is characterized by functional changes in the airways. The lung-gut axis and gut microbiota (GM) have been linked to the pathophysiology of airway diseases. Regarding COPD, studies have shown that GM alterations could be related the stages of this disease. However, the relationship between GM and clinical, biochemical and immunological parameters in patients with COPD are not well understood. The aim of this study was to compare the relative abundance of specific groups of beneficial gut bacteria between COPD patients and healthy controls (CTLs) in order to evaluate relationships with metabolic and inflammatory markers in COPD. METHODS We included 16 stable COPD patients and 16 healthy volunteer CTLs. The relative abundances of Bifidobacterium spp. (Bf) and Akkermansia muciniphila (Akk) bacteria and the Bacteroidetes and Firmicutes phyla were assessed by qPCR. Pulmonary function was evaluated by spirometry, biochemical parameters by colorimetric methods and plasma cytokine levels by cytometric bead array analysis. RESULTS The Firmicutes/Bacteroides ratio was related to emergency hospital visits and six-minute walk test (6MWT) results. Furthermore, the relative abundance of Bf was associated with plasma concentrations of glucose, triglycerides, HDL-C and IL-10. In addition, Firmicutes levels and the Firmicutes/Bacteroidetes ratio were associated with the IL-12/IL-10 ratio, while Akk abundance was linked to IL-12 levels. CONCLUSIONS The present findings suggest that the abundance of beneficial bacteria in the GM could influence clinical presentation and immunoregulation in COPD.
Collapse
Affiliation(s)
- Fabine Correia Passos
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40170-110, Bahia, Brazil; (F.C.P.); (L.M.G.d.O.); (O.L.L.N.)
| | - Lucas Matheus Gonçalves de Oliveira
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40170-110, Bahia, Brazil; (F.C.P.); (L.M.G.d.O.); (O.L.L.N.)
| | - Fabíola Ramos Jesus
- Maternidade Climério de Oliveira (MCO/EBSERH), Universidade Federal da Bahia, Salvador 40055-150, Bahia, Brazil;
| | | | - Odilon Lobão Leal Neto
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40170-110, Bahia, Brazil; (F.C.P.); (L.M.G.d.O.); (O.L.L.N.)
| | - Margarida Célia Lima Costa Neves
- Unidade do Sistema Respiratório, Ambulatório Professor Francisco Magalhães Neto, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador 40110-200, Bahia, Brazil; (M.C.L.C.N.); (A.C.M.L.)
| | - Antônio Carlos Moreira Lemos
- Unidade do Sistema Respiratório, Ambulatório Professor Francisco Magalhães Neto, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador 40110-200, Bahia, Brazil; (M.C.L.C.N.); (A.C.M.L.)
| | - Gyselle Chrystina Baccan
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40170-110, Bahia, Brazil; (F.C.P.); (L.M.G.d.O.); (O.L.L.N.)
| |
Collapse
|
46
|
Chen Y, Xiao L, Zhou M, Zhang H. The microbiota: a crucial mediator in gut homeostasis and colonization resistance. Front Microbiol 2024; 15:1417864. [PMID: 39165572 PMCID: PMC11333231 DOI: 10.3389/fmicb.2024.1417864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
The gut microbiota is a complex and diverse community of microorganisms that colonizes the human gastrointestinal tract and influences various aspects of human health. These microbes are closely related to enteric infections. As a foreign entity for the host, commensal microbiota is restricted and regulated by the barrier and immune system in the gut and contributes to gut homeostasis. Commensals also effectively resist the colonization of pathogens and the overgrowth of indigenous pathobionts by utilizing a variety of mechanisms, while pathogens have developed strategies to subvert colonization resistance. Dysbiosis of the microbial community can lead to enteric infections. The microbiota acts as a pivotal mediator in establishing a harmonious mutualistic symbiosis with the host and shielding the host against pathogens. This review aims to provide a comprehensive overview of the mechanisms underlying host-microbiome and microbiome-pathogen interactions, highlighting the multi-faceted roles of the gut microbiota in preventing enteric infections. We also discuss the applications of manipulating the microbiota to treat infectious diseases in the gut.
Collapse
Affiliation(s)
- Yiding Chen
- Department of Gastroenterology, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Ling Xiao
- Department of Gastroenterology, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Min Zhou
- Department of Gastroenterology, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Hu Zhang
- Department of Gastroenterology, West China Tianfu Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Center for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
47
|
Goya-Jorge E, Gonza I, Bondue P, Druart G, Al-Chihab M, Boutaleb S, Douny C, Taminiau B, Daube G, Scippo ML, Thonart P, Delcenserie V. Unveiling the influence of a probiotic combination of Heyndrickxia coagulans and Lacticaseibacillus casei on healthy human gut microbiota using the TripleSHIME® system. Microbiol Res 2024; 285:127778. [PMID: 38823185 DOI: 10.1016/j.micres.2024.127778] [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: 10/15/2023] [Revised: 04/30/2024] [Accepted: 05/17/2024] [Indexed: 06/03/2024]
Abstract
Probiotics are host-friendly microorganisms that can have important health benefits in the human gut microbiota as dietary supplements. Maintaining a healthy gut microbial balance relies on the intricate interplay among the intestinal microbiota, metabolic activities, and the host's immune response. This study aims to explore if a mixture of Heyndrickxia coagulans [ATB-BCS-042] and Lacticaseibacillus casei [THT-030-401] promotes in vitro this balance in representative gut microbiota from healthy individuals using the Triple-SHIME® (Simulation of the Human Intestinal Microbial Ecosystem). Metataxonomic analysis of the intestinal microbes revealed that the probiotic mix was not causing important disruptions in the biodiversity or microbial composition of the three simulated microbiota. However, some targeted populations analyzed by qPCR were found to be disrupted at the end of the probiotic treatment or after one week of washout. Populations such as Cluster IV, Cluster XVIa, and Roseburia spp., were increased indicating a potential gut health-promoting butyrogenic effect of the probiotic supplementation. In two of the systems, bifidogenic effects were observed, while in the third, the treatment caused a decrease in bifidobacteria. For the health-detrimental biomarker Escherichia-Shigella, a mild decrease in all systems was observed in the proximal colon sections, but these genera were highly increased in the distal colon sections. By the end of the washout, Bacteroides-Prevotella was found consistently boosted, which could have inflammatory consequences in the intestinal context. Although the probiotics had minimal influence on most quantified metabolites, ammonia consistently decreased after one week of daily probiotic supplementation. In reporter gene assays, aryl hydrocarbon receptor (AhR) activation was favored by the metabolic output obtained from post-treatment periods. Exposure of a human intestinal cell model to fermentation supernatant obtained after probiotic supplementation induced a trend to decrease the mRNA expression of immunomodulatory cytokines (IL-6, IL-8). Overall, with some exceptions, a positive impact of H. coagulans and L. casei probiotic mix was observed in the three parallel experiments, despite inter-individual differences. This study might serve as an in vitro pipeline for the impact assessment of probiotic combinations on the human gut microbiota.
Collapse
Affiliation(s)
- Elizabeth Goya-Jorge
- Laboratory of Food Quality Management, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, Liège 4000, Belgium; Intestinal Regenerative Medicine Lab, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA.
| | - Irma Gonza
- Laboratory of Food Quality Management, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, Liège 4000, Belgium.
| | - Pauline Bondue
- Laboratory of Food Quality Management, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, Liège 4000, Belgium.
| | - Germain Druart
- Lacto Research sprl., Rue Herman Meganck 21, Gembloux-les Isnes 5032, Belgium.
| | - Mohamed Al-Chihab
- Lacto Research sprl., Rue Herman Meganck 21, Gembloux-les Isnes 5032, Belgium.
| | - Samiha Boutaleb
- Laboratory of Food Analysis, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, Liège 4000, Belgium.
| | - Caroline Douny
- Laboratory of Food Analysis, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, Liège 4000, Belgium.
| | - Bernard Taminiau
- Laboratory of Microbiology, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, Liège 4000, Belgium.
| | - Georges Daube
- Laboratory of Microbiology, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, Liège 4000, Belgium.
| | - Marie-Louise Scippo
- Laboratory of Food Analysis, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, Liège 4000, Belgium.
| | - Philippe Thonart
- Lacto Research sprl., Rue Herman Meganck 21, Gembloux-les Isnes 5032, Belgium.
| | - Véronique Delcenserie
- Laboratory of Food Quality Management, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, Liège 4000, Belgium.
| |
Collapse
|
48
|
Si HR, Wu K, Su J, Dong TY, Zhu Y, Li B, Chen Y, Li Y, Shi ZL, Zhou P. Individual virome analysis reveals the general co-infection of mammal-associated viruses with SARS-related coronaviruses in bats. Virol Sin 2024; 39:565-573. [PMID: 38945213 PMCID: PMC11401474 DOI: 10.1016/j.virs.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/19/2024] [Indexed: 07/02/2024] Open
Abstract
Bats are the natural reservoir hosts for SARS-related coronavirus (SARSr-CoV) and other highly pathogenic microorganisms. Therefore, it is conceivable that an individual bat may harbor multiple microbes. However, there is limited knowledge on the overall co-circulation of microorganisms in bats. Here, we conducted a 16-year monitoring of bat viruses in south and central China and identified 238 SARSr-CoV positive samples across nine bat species from ten provinces or administrative districts. Among these, 76 individual samples were selected for further metagenomics analysis. We found a complex microenvironment characterized by the general co-circulation of microbes from two different sources: mammal-associated viruses or environment-associated microbes. The later includes commensal bacteria, enterobacteria-related phages, and insect or fungal viruses of food origin. Results showed that 25% (19/76) of the samples contained at least one another mammal-associated virus, notably alphacoronaviruses (13/76) such as AlphaCoV/YN2012, HKU2-related CoV and AlphaCoV/Rf-HuB2013, along with viruses from other families. Notably, we observed three viruses co-circulating within a single bat, comprising two coronavirus species and one picornavirus. Our analysis also revealed the potential presence of pathogenic bacteria or fungi in bats. Furthermore, we obtained 25 viral genomes from the 76 bat SARSr-CoV positive samples, some of which formed new evolutionary lineages. Collectively, our study reveals the complex microenvironment of bat microbiome, facilitating deeper investigations into their pathogenic potential and the likelihood of cross-species transmission.
Collapse
Affiliation(s)
- Hao-Rui Si
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43000, China; University of Chinese Academy of Sciences, Beijing 100000, China
| | - Ke Wu
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou 510005, China
| | - Jia Su
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43000, China; University of Chinese Academy of Sciences, Beijing 100000, China
| | - Tian-Yi Dong
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43000, China; University of Chinese Academy of Sciences, Beijing 100000, China
| | - Yan Zhu
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43000, China
| | - Bei Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43000, China
| | - Ying Chen
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43000, China
| | - Yang Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43000, China
| | - Zheng-Li Shi
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43000, China; Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou 510005, China.
| | - Peng Zhou
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou 510005, China; State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical School, Guangzhou 510005, China.
| |
Collapse
|
49
|
Liu M, Li S, Cao S, Liu C, Han Y, Cheng J, Zhang S, Zhao J, Shi Y. Let food be your medicine - dietary fiber. Food Funct 2024; 15:7733-7756. [PMID: 38984439 DOI: 10.1039/d3fo05641d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Dietary fiber (DF) cannot be digested and absorbed by the digestive tract, nor can it provide the energy needed to be burned for metabolic activities. Therefore, from the 1950s to the 1980s, DF received little attention in nutrition studies. With in-depth research and developments in global nutrition, people have gradually paid attention to the fact that DF occupies an essential position in the structure of nutrition, and it can ensure the healthy development of human beings. As early as 390 B.C., the ancient Greek physician Hippocrates proposed, "Let your food be your medicine, and your medicine be your food". This concept has been more systematically validated in modern scientific research, with numerous epidemiological studies showing that the dietary intake of DF-rich foods such as whole grains, root vegetables, legumes, and fruits has the potential to regulate the balance of the gut microbiota and thereby prevent diseases. However, the crosstalk between different types of DF and the gut microbiota is quite complex, and the effects on the organism vary. In this paper, we discuss research on DF and the gut microbiota and related diseases, aiming to understand the relationship between all three better and provide a reference basis for the risk reduction of related diseases.
Collapse
Affiliation(s)
- Mengqi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Shouren Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Shixi Cao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Cong Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Yao Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Jiawen Cheng
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Shuhang Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
| | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas, USA
| | - Yinghua Shi
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China.
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
- Henan Forage Engineering Technology Research Center, Zhengzhou, Henan, 450002, China
| |
Collapse
|
50
|
Zancan V, Nasello M, Bigi R, Reniè R, Buscarinu MC, Mechelli R, Ristori G, Salvetti M, Bellucci G. Gut Microbiota Composition Is Causally Linked to Multiple Sclerosis: A Mendelian Randomization Analysis. Microorganisms 2024; 12:1476. [PMID: 39065244 PMCID: PMC11278727 DOI: 10.3390/microorganisms12071476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/26/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Accumulating evidence links the microbial communities inhabiting the gut to the pathophysiological processes underlying multiple sclerosis (MS). However, most studies on the microbiome in MS are correlative in nature, thus being at risk of confounding and reverse causality. Mendelian randomization (MR) analyses allow the estimation of the causal relationship between a risk factor and an outcome of interest using genetic variants as proxies for environmental exposures. Here, we performed a two-sample MR to assess the causality between the gut microbiome and MS. We extracted genetic instruments from summary statistics from three large genome-wide association studies (GWASs) on the gut microbiome (18,340, 8959, and 7738 subjects). The exposure data were derived from the latest GWAS on MS susceptibility (47,429 patients and 68,374 controls). We pinpointed several microbial strains whose abundance is linked with enhanced MS risk (Actinobacteria class, Bifidobacteriaceae family, Lactobacillus genus) or protection (Prevotella spp., Lachnospiranaceae genus, Negativibacillus genus). The largest risk effect was seen for Ruminococcus Torques (OR, 2.89, 95% C.I. 1.67-5, p = 1.51 × 10-4), while Akkermansia municiphila emerged as strongly protective (OR, 0.43, 95% C.I. 0.32-0.57, p = 1.37 × 10-8). Our findings support a causal relationship between the gut microbiome and MS susceptibility, reinforcing the relevance of the microbiome-gut-brain axis in disease etiology, opening wider perspectives on host-environmental interactions for MS prevention.
Collapse
Affiliation(s)
- Valeria Zancan
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Martina Nasello
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Rachele Bigi
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, 00179 Rome, Italy
| | - Roberta Reniè
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Chiara Buscarinu
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, 00179 Rome, Italy
| | - Rosella Mechelli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00163 Rome, Italy
- Department for the Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Giovanni Ristori
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, 00179 Rome, Italy
| | - Marco Salvetti
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Mediterraneo Neuromed, 86077 Pozzilli, Italy
| | - Gianmarco Bellucci
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|