1
|
Uchida A, Imai K, Miki R, Hamaguchi T, Nishiwaki H, Ito M, Ueyama J, Hattori S, Tano S, Fuma K, Matsuo S, Ushida T, Ohno K, Kajiyama H, Kotani T. Butyrate-producing bacteria in pregnancy maintenance: mitigating dysbiosis-induced preterm birth. J Transl Med 2025; 23:533. [PMID: 40355924 PMCID: PMC12067704 DOI: 10.1186/s12967-025-06534-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Preterm birth (PTB) is a major contributor to neonatal morbidity, mortality, and long-term health complications. Despite advances in perinatal care, PTB rates remain high, and its multifactorial etiology is not fully understood. Increasing evidence suggests that maternal gut microbiota plays a critical role in pregnancy maintenance, potentially through modulation of immune responses. However, the underlying causal mechanisms remain unclear. We hypothesized that dysbiosis disrupts immune tolerance and promotes PTB, and that butyrate (short-chain fatty acid produced by specific gut bacteria) may counteract this effect by enhancing regulatory T cell (Treg)-mediated immune regulation. METHODS We established a dysbiosis-induced PTB mouse model using vancomycin treatment combined with subclinical immune activation via anti-CD3ε antibody. Pregnant mice were fed either a standard or butyrate-enriched diet. Outcomes included gestational length, PTB incidence, live pup rates, and Treg cell levels assessed by flow cytometry. Parallelly, 16S rRNA gene sequencing was performed on fecal samples from 32 pregnant women to compare gut microbial composition between spontaneous PTB and term birth groups. Multivariate logistic regression and correlation analyses were conducted to assess associations with gestational outcomes. RESULTS Vancomycin-induced dysbiosis in mice significantly reduced Treg cell populations and increased PTB rates (43.3% in dysbiosis vs. 0% in controls; p < 0.05), while butyrate supplementation reduced PTB incidence (p = 0.03), prolonged gestation (p = 0.01), and restored Treg counts (p < 0.001). In human samples, significant reductions in Lachnospiraceae and Ruminococcaceae, representative butyrate-producing bacteria, were seen in PTB cases. Their combined abundance was independently associated with sPTB risk (p = 0.019) and positively correlated with gestational age (r = 0.59, p < 0.001). CONCLUSIONS Our findings demonstrate that maternal dysbiosis increases PTB risk via impaired immune tolerance, and that butyrate supplementation effectively reverses this effect in vivo. Human data support the translational relevance of butyrate-producing microbiota in pregnancy maintenance. These results highlight butyrate as a promising target for dietary interventions aimed at reducing PTB incidence by restoring immune homeostasis. Trial registration Not applicable.
Collapse
Affiliation(s)
- Azusa Uchida
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kenji Imai
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Rika Miki
- Maternal Fetal Health Laboratory, Research Institute, Nozaki Tokushukai Hospital, Daito, Osaka, Japan
- Laboratory of Bell Research Center‑Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomonari Hamaguchi
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Nishiwaki
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun Ueyama
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satomi Hattori
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Sho Tano
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kazuya Fuma
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Seiko Matsuo
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Takafumi Ushida
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomomi Kotani
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
2
|
Wang G, Ma T, Liu R, Gu H, Zhou ZY, Wan Z. Comparisons of metabolites and gut microbiota profiles for both young and middle-aged APPSwe/PS1De9 mice. Neuroscience 2025; 577:54-63. [PMID: 40355072 DOI: 10.1016/j.neuroscience.2025.04.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 04/24/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
The research focused on exploring the differences and relationships between gut microbiota and metabolites at various stages of Alzheimer's disease (AD), specifically using APP/PS1 mice at the ages of 6 months and 10 months. To assess metabolites in serum and cortex, and to evaluate gut microbiota profiles in cecal content, UPLC-MS/MS and 16S rRNA sequencing techniques were utilized, respectively. Findings indicated that, in comparison to younger mice, serum concentrations of L-Leucine, thymine, and Glucosamine 6-phosphate were lower, whereas levels of Sorbitol and Palmitic acid were higher. Furthermore, measurements of the ACE and Chao1 indices significantly declined in the older cohort. At the phylum level, the relative abundance of Bacteroidetes showed a decline, while there was an increase in Actinobacteria and TM7 bacteria among the middle-aged subjects. The novelty of this study is we found there were notable alterations in both gut microbiota and metabolites within serum and cortex when comparing young and older APP/PS1 mice, emphasizing the important connections between metabolites and gut microbiota throughout the progression of AD. These results indicate that manipulating metabolites and gut flora may serve as a vital strategy for the prevention and management of AD.
Collapse
Affiliation(s)
- Guiping Wang
- Laboratory Animal Center, Medical College of Soochow University, 199 Ren'ai Road, Suzhou, China
| | - Tongtong Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou, China
| | - Ruitong Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou, China
| | - Huiwen Gu
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou, China
| | - Zheng-Yu Zhou
- Laboratory Animal Center, Medical College of Soochow University, 199 Ren'ai Road, Suzhou, China.
| | - Zhongxiao Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou, China.
| |
Collapse
|
3
|
Brasiel PGDA, Dutra Medeiros J, Costa de Almeida T, Teodoro de Souza C, de Cássia Ávila Alpino G, Barbosa Ferreira Machado A, Dutra Luquetti SCP. Preventive effects of kefir on colon tumor development in Wistar rats: gut microbiota critical role. J Dev Orig Health Dis 2025; 16:e5. [PMID: 39868980 DOI: 10.1017/s2040174424000461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
To clarify the effects of kefir in critical periods of development in adult diseases, we study the effects of kefir intake during early life on gut microbiota and prevention of colorectal carcinogenesis in adulthood. Lactating Wistar rats were divided into three groups: control (C), kefir lactation (KL), and kefir puberty (KP) groups. The C and KP groups received 1 mL of water/day; KL dams received kefir milk daily (108 CFU/mL) during lactation. After weaning (postnatal day 21), KP pups received kefir treatment until 60 days. At 67 days old, colorectal carcinogenesis was induced through intraperitoneal injection of 1, 2-dimethylhydrazine. The gut microbiota composition were analyzed by 16S rRNA gene sequencing and DESeq2 (differential abundance method), revealing significant differences in bacterial abundances between the kefir consumption periods. Maternal kefir intake strong anticancer power, suppressed tumors in adult offspring and reduced the relative risk of offspring tumor development. The gut microbiota in cecal samples of the KL group was enriched with Lactobacillus, Romboutsia, and Blautia. In contrast, control animals were enriched with Acinetobacter. The administration of kefir during critical periods of development, with emphasis on lactation, affected the gut microbial community structure to promote host benefits. Pearson analysis indicated positive correlation between tumor number with IL-1 levels. Therefore, the probiotic fermented food intake in early life may be effective as chemopreventive potential against colon tumor development, especially in lactation period.
Collapse
Affiliation(s)
| | - Julliane Dutra Medeiros
- Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Thaís Costa de Almeida
- Department of Nutrition, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Claudio Teodoro de Souza
- Department of Clinical Medicine, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | | | | | | |
Collapse
|
4
|
Schwieters A, Ahmer BMM. Role of the LuxR solo, SdiA, in eavesdropping on foreign bacteria. FEMS Microbiol Rev 2025; 49:fuaf015. [PMID: 40240290 DOI: 10.1093/femsre/fuaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/30/2025] [Accepted: 04/15/2025] [Indexed: 04/18/2025] Open
Abstract
Bacteria can cooperate by coordinating their gene expression through the production, release, and detection of small molecules, a phenomenon known as quorum sensing (QS). One type of QS commonly found in Gram-negative bacteria utilizes a LuxI-type enzyme to produce a signaling molecule of the N-acyl-homoserine lactone (AHL) family, and a transcription factor of the LuxR family to detect and respond to the AHL. In a subset of Enterobacteriaceae, including Escherichia coli and Salmonella, no LuxI family member is present and no AHLs are synthesized. However, they encode a LuxR family member, SdiA, that is used to detect the QS molecules of other bacterial species, a behavior known as eavesdropping. Despite significant research on the topic, the overall role of SdiA-mediated eavesdropping in these bacteria remains unclear. In this review, we discuss the phenotypes and regulons of SdiA in the Enterobacteriaceae.
Collapse
Affiliation(s)
- Andrew Schwieters
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, United States
| | - Brian M M Ahmer
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, United States
| |
Collapse
|
5
|
Li J, Gao F, Li R, Chen Z, Chen G, Fan P, Du G. Endoscopic surgery affects the gut microbiota and its metabolism in breast cancer patients. Front Microbiol 2025; 15:1481582. [PMID: 39839115 PMCID: PMC11747589 DOI: 10.3389/fmicb.2024.1481582] [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: 08/19/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Background Despite the advantages of endoscopic surgery in reducing trauma and enhancing recovery for breast cancer patients, its impact on gut microbiota, which is crucial for health and estrogen metabolism, remains unclear. Further investigation is necessary to fully understand this impact and its implications. Materials and methods Between June and December 2022, fecal samples were collected from 20 patients who underwent endoscopic surgery. The gut microbiota composition was determined using 16S rRNA sequencing, while the metabolites were analyzed through liquid chromatography-tandem mass spectrometry (LC-MS/MS). Bioinformatics and statistical analyses were employed to identify significant alterations in microbial taxa abundance and to assess intergroup differences. These analyses included t-tests for pairwise comparisons, one-way ANOVA for multiple group comparisons, and chi-square tests for categorical data analysis. Results Endoscopic surgery in breast cancer patients subtly changed gut microbiota diversity and composition. Post-surgery, there was a reduction in Lachnospiraceae, Monoglobaceae and Firmicutes to Bacteroides ratios. Shifts in metabolites were also observed, the changed metabolites impacted pathways such as primary bile biosynthesis and Ascorbate and aldarate metabolism, with PE(PGD1/18:1(9Z)) identified as a key differential metabolite that increased post-surgery. Azasetron, tyramine glucuronide, DL-DOPA, phthalide, acetophenazine, aciclovir, creatinine bicarbonate, and 4-oxo-L-proline being associated with distinct bacterial taxa. Conclusion Breast cancer patients undergoing endoscopic surgery experience a shift in their gut microbiota and metabolic profiles. Therefore, postoperative management, with a particular focus on the adjustment of the gut microbiota, is crucial for enhancing patient recovery and health outcomes.
Collapse
Affiliation(s)
- Jingtai Li
- The First Clinical School of Hainan Medical University, Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Fangfang Gao
- The First Clinical School of Hainan Medical University, Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Runwei Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Zhilin Chen
- The First Clinical School of Hainan Medical University, Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Guoping Chen
- The First Clinical School of Hainan Medical University, Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Pingming Fan
- The First Clinical School of Hainan Medical University, Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Guankui Du
- The First Clinical School of Hainan Medical University, Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
- Department of Biochemistry and Molecular Biology, Hainan Medical University, Haikou, China
| |
Collapse
|
6
|
Kim Y, Kokkinias K, Sabag-Daigle A, Leleiwi I, Borton M, Shaffer M, Baniasad M, Daly R, Ahmer BMM, Wrighton KC, Wysocki VH. Time-Resolved Multiomics Illustrates Host and Gut Microbe Interactions during Salmonella Infection. J Proteome Res 2024; 23:4864-4877. [PMID: 39374136 DOI: 10.1021/acs.jproteome.4c00172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Salmonella infection, also known as Salmonellosis, is one of the most common food-borne illnesses. Salmonella infection can trigger host defensive functions, including an inflammatory response. The provoked-host inflammatory response has a significant impact on the bacterial population in the gut. In addition, Salmonella competes with other gut microorganisms for survival and growth within the host. Compositional and functional alterations in gut bacteria occur because of the host immunological response and competition between Salmonella and the gut microbiome. Host variation and the inherent complexity of the gut microbial community make understanding commensal and pathogen interactions particularly difficult during a Salmonella infection. Here, we present metabolomics and lipidomics analyses along with the 16S rRNA sequence analysis, revealing a comprehensive view of the metabolic interactions between the host and gut microbiota during Salmonella infection in a CBA/J mouse model. We found that different metabolic pathways were altered over the four investigated time points of Salmonella infection (days -2, +2, +6, and +13). Furthermore, metatranscriptomics analysis integrated with metabolomics and lipidomics analysis facilitated an understanding of the heterogeneous response of mice, depending on the degree of dysbiosis.
Collapse
Affiliation(s)
- Yongseok Kim
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
| | - Katherine Kokkinias
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Anice Sabag-Daigle
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ikaia Leleiwi
- Department of Cell & Molecular Biology, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Mikayla Borton
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Michael Shaffer
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Maryam Baniasad
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
| | - Rebecca Daly
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Brian M M Ahmer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio 43210, United States
| | - Kelly C Wrighton
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Vicki H Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
7
|
Meng S, Xing S, Xu H, Li J, Jiang Y, He H, Cai H, Li M. Integrated analysis of intestinal microbial community and muscle transcriptome profile in rabbits. Anim Biotechnol 2024; 35:2387015. [PMID: 39145993 DOI: 10.1080/10495398.2024.2387015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Intestinal microbial community plays an important part in maintaining health and skeletal muscle development in livestock. This study is the first of its kind in the world. In order to better understand the relationship between gut microbiota and gene expression in skeletal muscle of rabbits, caecum contents and longissimus dorsi tissues of rabbits at 0 d (S1), 35 d (S2) and 70d (S3) were collected and subjected for 16S rRNA sequencing and transcriptome sequencing. Our results showed that, among three groups of rabbits, Firmicutes and Bacteroidetes were the dominant phyla at the phylum level, while Akmansia, Bacteroides and Ruminobacter were the dominant genera at the genus level, and the relative abundance of Akmansia and Bacteroides increased firstly and then decreased from 0 d to 70 d. By analyzing the transcriptome sequencing data, we identified 2866, 2446 and 4541 differentially expressed genes (DEGs) in S1 vs S2, S2 vs S3 and S1 vs S3 groups, respectively. Finally, we performed correlation analysis between gut microbiota and the expression levels of muscle development-related genes of rabbits at 0 d and 70 d. Compared with 0 day old rabbits, in 70 day old rabbits Acinetobacter and Cronbacter with decreased abundance, and Ruminococcaceae_UCG-014 and Ruminococcus_1 with increase abundance is beneficial to caecum health in rabbits. These results will lay a foundation for further re-searches about the relationship between caecum microflora and muscle development in rabbits.
Collapse
Affiliation(s)
- Shengbo Meng
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, P.R. China
| | - Shanshan Xing
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, P.R. China
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, P.R. China
| | - Jing Li
- Animal Health Supervision Institute of Biyang, Henan, P.R. China
| | - Yixuan Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, P.R. China
| | - Hui He
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, P.R. China
| | - Hanfang Cai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, P.R. China
| | - Ming Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, P.R. China
| |
Collapse
|
8
|
Kokkinias K, Sabag-Daigle A, Kim Y, Leleiwi I, Shaffer M, Kevorkian R, Daly RA, Wysocki VH, Borton MA, Ahmer BMM, Wrighton KC. Time-resolved multi-omics reveals diverse metabolic strategies of Salmonella during diet-induced inflammation. mSphere 2024; 9:e0053424. [PMID: 39254340 PMCID: PMC11520297 DOI: 10.1128/msphere.00534-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 07/22/2024] [Indexed: 09/11/2024] Open
Abstract
With a rise in antibiotic resistance and chronic infection, the metabolic response of Salmonella enterica serovar Typhimurium to various dietary conditions over time remains an understudied avenue for novel, targeted therapeutics. Elucidating how enteric pathogens respond to dietary variation not only helps us decipher the metabolic strategies leveraged for expansion but also assists in proposing targets for therapeutic interventions. In this study, we use a multi-omics approach to identify the metabolic response of Salmonella enterica serovar Typhimurium in mice on both a fibrous diet and high-fat diet over time. When comparing Salmonella gene expression between diets, we found a preferential use of respiratory electron acceptors consistent with increased inflammation in high-fat diet mice. Looking at the high-fat diet over the course of infection, we noticed heterogeneity in samples based on Salmonella ribosomal activity, which is separated into three infection phases: early, peak, and late. We identified key respiratory, carbon, and pathogenesis gene expressions descriptive of each phase. Surprisingly, we identified genes associated with host cell entry expressed throughout infection, suggesting subpopulations of Salmonella or stress-induced dysregulation. Collectively, these results highlight not only the sensitivity of Salmonella to its environment but also identify phase-specific genes that may be used as therapeutic targets to reduce infection.IMPORTANCEIdentifying novel therapeutic strategies for Salmonella infection that occur in relevant diets and over time is needed with the rise of antibiotic resistance and global shifts toward Western diets that are high in fat and low in fiber. Mice on a high-fat diet are more inflamed compared to those on a fibrous diet, creating an environment that results in more favorable energy generation for Salmonella. We observed differential gene expression across infection phases in mice over time on a high-fat diet. Together, these findings reveal the metabolic tuning of Salmonella to dietary and temporal perturbations. Research like this, which explores the dimensions of pathogen metabolic plasticity, can pave the way for rationally designed strategies to control disease.
Collapse
Affiliation(s)
- Katherine Kokkinias
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Anice Sabag-Daigle
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Yongseok Kim
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Ikaia Leleiwi
- Department of Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA
| | - Michael Shaffer
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Richard Kevorkian
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Rebecca A. Daly
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Vicki H. Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Mikayla A. Borton
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Brian M. M. Ahmer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Kelly C. Wrighton
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
- Department of Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
9
|
Schwieters A, Ahmer BMM. Identification of new SdiA regulon members of Escherichia coli, Enterobacter cloacae, and Salmonella enterica serovars Typhimurium and Typhi. Microbiol Spectr 2024; 12:e0192924. [PMID: 39436139 PMCID: PMC11619404 DOI: 10.1128/spectrum.01929-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/16/2024] [Indexed: 10/23/2024] Open
Abstract
Bacteria can coordinate behavior in response to population density through the production, release, and detection of small molecules, a phenomenon known as quorum sensing. Salmonella enterica is among a group of Enterobacteriaceae that can detect signaling molecules of the N-acyl homoserine lactone (AHL) type but lack the ability to produce them. The AHLs are detected by the LuxR-type transcription factor, SdiA. This enables a behavior known as eavesdropping, where organisms can sense the signaling molecules of other species of bacteria. The role of SdiA remains largely unknown. Here, we use RNA-seq to more completely identify the sdiA regulons of two clinically significant serovars of Salmonella enterica: Typhimurium and Typhi. We find that their sdiA regulons are largely conserved despite the significant differences in pathogenic strategy and host range of these two serovars. Previous studies identified sdiA-regulated genes in Escherichia coli and Enterobacter cloacae, but there is surprisingly no overlap in regulon membership between the different species. This led us to individually test orthologs of each regulon member in the other species and determine that there is indeed some overlap. Unfortunately, the functions of most sdiA-regulated genes are unknown, with the overall function of eavesdropping in these organisms remaining unclear. IMPORTANCE Many bacterial species detect their own population density through the production, release, and detection of small molecules (quorum sensing). Salmonella and other Enterobacteriaceae have a modified system that detects the N-acyl-homoserine lactones of other bacteria through the solo quorum sensing receptor SdiA, a behavior known as eavesdropping. The roles of sdiA-dependent eavesdropping in the lifecycles of these bacteria are unknown. In this study, we identify sdiA-dependent transcriptional responses in two clinically relevant serovars of Salmonella, Typhimurium and Typhi, and note that their responses are partially conserved. We also demonstrate for the first time that sdiA-dependent regulation of genes is partially conserved in Enterobacter cloacae and Escherichia coli as well, indicating a degree of commonality in eavesdropping among the Enterobacteriaceae.
Collapse
Affiliation(s)
- Andrew Schwieters
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Brian M. M. Ahmer
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
10
|
Chen S, Du F, Shang K, Chen H, Guo R, Liao C, Jia Y, Yu Z, Li J, Zhang C, Ding K. Colonization Mediated by T6SS-ClpV Disrupts Host Gut Microbiota and Enhances Virulence of Salmonella enterica serovar Typhimurium. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19155-19166. [PMID: 39161106 DOI: 10.1021/acs.jafc.4c03735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is a common foodborne enteric pathogen that infects humans or mammals and colonizes the intestinal tract primarily by invading the host following ingestion. Meanwhile, ClpV is a core secreted protein of the bacterial type VI secretion system (T6SS). Because elucidating ClpV's role in the pathogenesis of T6SS is pivotal for revealing the virulence mechanism of Salmonella, in our study, clpV gene deletion mutants were constructed using a λ-red-based recombination system, and the effect of clpV mutation on SL1344's pathogenicity was examined in terms of stress resistance, motility, cytokine secretion, gut microbiota, and a BALB/c mouse model. Among the results, ClpV affected SL1344's motility and was also involved in cell invasion, adhesion, and intracellular survival in the MDBK cell model but did not affect invasion or intracellular survival in the RAW264.7 cell model. Moreover, clpV gene deletion significantly reduced the transcription levels of GBP2b, IFNB1, IL-6, NLRP3, NOS2, and TNF-α proinflammatory factor levels but significantly increased transcription levels of IL-4 and IL-10 anti-inflammatory factors. Last, ClpV appeared to closely relate to the pathogenicity of S. Typhimurium in vivo, which can change the gut environment and cause dysbiosis of gut microbiota. Our findings elucidate the functions of ClpV in S. Typhimurium and illustrating interactions between T6SS and gut microbiota help to clarify the mechanisms of the pathogenesis of foodborne diseases.
Collapse
Affiliation(s)
- Songbiao Chen
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471023, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan 450000, China
| | - Fuxi Du
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471023, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Ke Shang
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471023, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Huimin Chen
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471023, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Rongxian Guo
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471023, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Chengshui Liao
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471023, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan 450000, China
| | - Yanyan Jia
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471023, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Zuhua Yu
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471023, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Jing Li
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471023, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Chunjie Zhang
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471023, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Ke Ding
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471023, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan 450000, China
| |
Collapse
|
11
|
Ma B, Wang D, Chen X, Wang Q, Zhang T, Wen R, Yang M, Li C, Lei C, Wang H. Dietary α-linolenic acid supplementation enhances resistance to Salmonella Typhimurium challenge in chickens by altering the intestinal mucosal barrier integrity and cecal microbes. Microbiol Res 2024; 285:127773. [PMID: 38833830 DOI: 10.1016/j.micres.2024.127773] [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: 04/11/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 06/06/2024]
Abstract
Salmonella is an important foodborne pathogen. Given the ban on the use of antibiotics during the egg-laying period in China, finding safe and effective alternatives to antibiotics to reduce Salmonella enterica subsp. enterica serovar Typhimurium (S. Typhimurium) infections in chickens is essential for the prevention and control of this pathogen and the protection of human health. Numerous studies have shown that unsaturated fatty acids have a positive effect on intestinal inflammation and resistance to infection by intestinal pathogens. Here we investigated the protective effect of α-linolenic acid (ALA) against S. Typhimurium infection in chickens and further explored its mechanism of action. We added different proportions of ALA to the feed and observed the effect of ALA on S. Typhimurium colonization using metagenomic sequencing technology and physiological index measurements. The role of gut flora on S. Typhimurium colonization was subsequently verified by fecal microbiota transplantation (FMT). We found that ALA protects chickens from S. Typhimurium infection by reducing intestinal inflammation through remodeling the gut microbiota, up-regulating the expression of ileocecal barrier-related genes, and maintaining the integrity of the intestinal epithelium. Our data suggest that supplementation of feed with ALA may be an effective strategy to alleviate S. Typhimurium infection in chickens.
Collapse
Affiliation(s)
- Boheng Ma
- College of Life Sciences, Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, People's Republic of China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, People's Republic of China
| | - De Wang
- College of Life Sciences, Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, People's Republic of China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, People's Republic of China
| | - Xuan Chen
- College of Life Sciences, Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, People's Republic of China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, People's Republic of China
| | - Qin Wang
- College of Life Sciences, Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, People's Republic of China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, People's Republic of China
| | - Tiejun Zhang
- College of Life Sciences, Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, People's Republic of China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, People's Republic of China
| | - Renqiao Wen
- College of Life Sciences, Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, People's Republic of China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, People's Republic of China
| | - Ming Yang
- College of Life Sciences, Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, People's Republic of China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, People's Republic of China
| | - Cui Li
- College of Life Sciences, Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, People's Republic of China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, People's Republic of China
| | - Changwei Lei
- College of Life Sciences, Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, People's Republic of China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, People's Republic of China.
| | - Hongning Wang
- College of Life Sciences, Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, People's Republic of China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, People's Republic of China.
| |
Collapse
|
12
|
Lu G, Gao D, Liu Y, Yu X, Jiang W, Lv Z. Early and long-term responses of intestinal microbiota and metabolites to 131I treatment in differentiated thyroid cancer patients. BMC Med 2024; 22:300. [PMID: 39020393 PMCID: PMC11256643 DOI: 10.1186/s12916-024-03528-3] [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: 02/06/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Multiple high doses of 131I therapy in patients with differentiated thyroid cancer (DTC) might disrupt the balance of gut microbiota and metabolites. This study aimed to investigate the alterations of intestinal bacteria and metabolism over two courses of 131I therapy, explore the interactions, and construct diagnostic models reflecting enteric microecology based on 131I therapy. METHODS A total of 81 patients were recruited for the first 131I therapy (131I-1st), among whom 16 received a second course (131I-2nd) after half a year. Fecal samples were collected 1 day before (Pre-131I-1st/2nd) and 3 days after (Post-131I-1st/2nd) 131I therapy for microbiome (16S rRNA gene sequencing) and metabolomic (LC-MS/MS) analyses. RESULTS A total of six microbial genera and 11 fecal metabolites enriched in three pathways were identified to show significant differences between Pre-131I-1st and other groups throughout the two courses of 131I treatment. In the Post-131I-1st group, the beneficial bacteria Bifidobacterium, Lachnoclostridium, uncultured_bacterium_f_Lachnospiraceae, and Lachnospiraceae_UCG004 were abundant and the radiation-sensitive pathways of linoleic acid (LA), arachidonic acid, and tryptophan metabolism were inhibited compared with the Pre-131I-1st group. Compared with the Pre-131I-1st group, the Pre-131I-2nd group exhibited a reduced diversity of flora and differentially expressed metabolites, with a low abundance of beneficial bacteria and dysregulated radiation-sensitive pathways. However, less significant differences in microbiota and metabolites were found between the Pre/Post-131I-2nd groups compared with those between the Pre/Post-131I-1st groups. A complex co-occurrence was observed between 6 genera and 11 metabolites, with Lachnoclostridium, Lachnospiraceae_UCG004, Escherichia-Shigella, and LA-related metabolites contributing the most. Furthermore, combined diagnostic models of charactered bacteria and metabolites answered well in the early, long-term, and dose-dependent responses for 131I therapy. CONCLUSIONS Different stages of 131I therapy exert various effects on gut microecology, which play an essential role in regulating radiotoxicity and predicting the therapeutic response.
Collapse
Affiliation(s)
- Ganghua Lu
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Institute of Clinical Mass Spectrometry Applied Research Center, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Dingwei Gao
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Institute of Clinical Mass Spectrometry Applied Research Center, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Yixian Liu
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaqing Yu
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Institute of Clinical Mass Spectrometry Applied Research Center, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| | - Wen Jiang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Institute of Clinical Mass Spectrometry Applied Research Center, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Zhongwei Lv
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Institute of Clinical Mass Spectrometry Applied Research Center, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 200003, China.
| |
Collapse
|
13
|
Guo J, Jia P, Gu Z, Tang W, Wang A, Sun Y, Li Z. Altered gut microbiota and metabolite profiles provide clues in understanding resistant hypertension. J Hypertens 2024; 42:1212-1225. [PMID: 38690877 DOI: 10.1097/hjh.0000000000003716] [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: 05/03/2024]
Abstract
BACKGROUND Resistant hypertension is a severe phenotype in hypertension that may be driven by interactions between genetic and environmental factors. Specific changes in gut microbiota and metabolites have been shown to influence cardiovascular disease progression. However, microbial and metabolomic changes associated with resistant hypertension remain elusive. METHODS In this study, the gut microbiome of 30 participants with resistant hypertension, 30 with controlled hypertension, and 30 nonhypertension was characterized using 16S rRNA amplicon sequencing. In addition, the serum metabolome of the same population was assessed by untargeted metabolomics. RESULTS The alpha diversity of microbiome in the resistant hypertension decreased, and changes were also observed in the composition of the gut microbiota. The resistant hypertension group was characterized by elevated levels of Actinobacteitia and Proteobacteria. Twenty-three genera were found to have significantly different abundances between resistant hypertension and controlled hypertension, as well as 55 genera with significantly different abundances between resistant hypertension and nonhypertension. Compared with the controlled hypertension group, the genera Rothia and Sharpea in resistant hypertension were more abundant. Compared with the nonhypertension group, the genera Escherichia-Shigella , Lactobacillus , Enterococcus were more abundant. Untargeted metabolomics provided distinctly different serum metabolic profiles for the three groups and identified a range of differential metabolites. These metabolites were mainly associated with the pathway of glycerophospholipid metabolism. Furthermore, correlation analysis provided evidence of new interactions between gut microbiota and metabolites in the resistant hypertension. CONCLUSION In conclusion, our study provides a comprehensive understanding of the resistant hypertension gut microbiota and metabolites, suggesting that treatment resistance in resistant hypertension patients may be related to the gut microbiota and serum metabolites.
Collapse
Affiliation(s)
- Jiuqi Guo
- Department of Cardiology, the First Hospital of China Medical University, Shenyang
| | - Pengyu Jia
- Department of Cardiology, the First Hospital of China Medical University, Shenyang
| | - Zhilin Gu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang
| | - Wenyi Tang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang
| | - Ai Wang
- Department of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang
| | - Zhao Li
- Department of Cardiology, the First Hospital of China Medical University, Shenyang
| |
Collapse
|
14
|
Feng Z, Liao M, Bai J, Li Y, Chen Y, Zhang L, Guo X, Li L, Zhang L. Exploring the causal relationship between gut microbiota and multiple myeloma risk based on Mendelian randomization and biological annotation. Front Microbiol 2024; 15:1310444. [PMID: 38410384 PMCID: PMC10895040 DOI: 10.3389/fmicb.2024.1310444] [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: 10/09/2023] [Accepted: 01/15/2024] [Indexed: 02/28/2024] Open
Abstract
Introduction The microbial genome-wide association studies (mbGWAS) have highlighted significant host-microbiome interactions based on microbiome heritability. However, establishing causal relationships between particular microbiota and multiple myeloma (MM) remains challenging due to limited sample sizes. Methods Gut microbiota data from a GWAS with 18,340 participants and MM summary statistics from 456,348 individuals. The inverse variance-weighted (IVW) method was used as the main bidirectional Mendelian randomization (MR) analysis. To assess the robustness of our results, we further performed supplementary analyses, including MR pleiotropy residual sum and outlier (MR-PRESSO) test, MR-Egger, Weighted median, Simple mode, and Weighted mode. Moreover, a backward MR analysis was conducted to investigate the potential for reverse causation. Finally, gene and gene-set-based analyses were then conducted to explore the common biological factors connecting gut microbiota and MM. Results We discovered that 10 gut microbial taxa were causally related to MM risk. Among them, family Acidaminococcaceae, Bacteroidales family S24-7, family Porphyromonadaceae, genus Eubacterium ruminantium group, genus Parabacteroides, and genus Turicibacter were positively correlated with MM. Conversely, class Verrucomicrobia, family Verrucomicrobiaceae, genus Akkermansia, and order Verrucomicrobiales were negatively correlated with MM. The heterogeneity test revealed no Heterogeneity. MR-Egger and MR-PRESSO tests showed no significant horizontal pleiotropy. Importantly, leave-one-out analysis confirmed the robustness of MR results. In the backward MR analysis, no statistically significant associations were discovered between MM and 10 gut microbiota taxa. Lastly, we identified novel host-microbiome shared genes (AUTS2, CDK2, ERBB3, IKZF4, PMEL, SUOX, and RAB5B) that are associated with immunoregulation and prognosis in MM through biological annotation. Discussion Overall, this study provides evidence supporting a potential causal relationship between gut microbiota and MM risk, while also revealing novel host-microbiome shared genes relevant to MM immunoregulation and clinical prognosis.
Collapse
Affiliation(s)
- Zuxi Feng
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Minjing Liao
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jun Bai
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Yanhong Li
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Yue Chen
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Li Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Xuege Guo
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lijuan Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Liansheng Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
15
|
Mishra SP, Jain S, Wang B, Wang S, Miller BC, Lee JY, Borlongan CV, Jiang L, Pollak J, Taraphder S, Layden BT, Rane SG, Yadav H. Abnormalities in microbiota/butyrate/FFAR3 signaling in aging gut impair brain function. JCI Insight 2024; 9:e168443. [PMID: 38329121 PMCID: PMC10967378 DOI: 10.1172/jci.insight.168443] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 12/08/2023] [Indexed: 02/09/2024] Open
Abstract
Aging-related abnormalities in gut microbiota are associated with cognitive decline, depression, and anxiety, but underlying mechanisms remain unstudied. Here, our study demonstrated that transplanting old gut microbiota to young mice induced inflammation in the gut and brain coupled with cognitive decline, depression, and anxiety. We observed diminished mucin formation and increased gut permeability ("leaky gut") with a reduction in beneficial metabolites like butyrate because of decline in butyrate-producing bacteria in the aged gut microbiota. This led to suppressed expression of butyrate receptors, free fatty acid receptors 2 and 3 (FFAR2/3). Administering butyrate alleviated inflammation, restored mucin expression and gut barriers, and corrected brain dysfunction. Furthermore, young mice with intestine-specific loss of FFAR2/3 exhibited gut and brain abnormalities akin to those in older mice. Our results demonstrate that reduced butyrate-producing bacteria in aged gut microbiota result in low butyrate levels and reduced FFAR2/3 signaling, leading to suppressed mucin formation that increases gut permeability, inflammation, and brain abnormalities. These findings underscore the significance of butyrate-FFAR2/3 agonism as a potential strategy to mitigate aged gut microbiota-induced detrimental effects on gut and brain health in older adults.
Collapse
Affiliation(s)
- Sidharth P. Mishra
- USF Center for Microbiome Research
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
| | - Shalini Jain
- USF Center for Microbiome Research
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
| | - Bo Wang
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, Florida, USA
| | - Shaohua Wang
- USF Center for Microbiome Research
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
| | - Brandi C. Miller
- USF Center for Microbiome Research
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
| | - Jea Y. Lee
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
| | - Cesar V. Borlongan
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
| | - Lin Jiang
- Natural Sciences Division, New College of Florida, Sarasota, Florida, USA
| | - Julie Pollak
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, Florida, USA
| | - Subhash Taraphder
- Department of Animal Genetics and Breeding, West Bengal University of Animal & Fishery Sciences, Kolkata, India
| | - Brian T. Layden
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Sushil G. Rane
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Hariom Yadav
- USF Center for Microbiome Research
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, USF Morsani College of Medicine, Tampa, Florida, USA
| |
Collapse
|
16
|
Sepúlveda-Alfaro J, Catalán EA, Vallejos OP, Ramos-Tapia I, Madrid-Muñoz C, Mendoza-León MJ, Suazo ID, Rivera-Asin E, Silva PH, Alvarez-Mardones O, Castillo-Godoy DP, Riedel CA, Schinnerling K, Ugalde JA, Soto JA, Bueno SM, Kalergis AM, Melo-Gonzalez F. Human metapneumovirus respiratory infection affects both innate and adaptive intestinal immunity. Front Immunol 2024; 15:1330209. [PMID: 38404579 PMCID: PMC10884822 DOI: 10.3389/fimmu.2024.1330209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/15/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Respiratory infections are one of the leading causes of morbidity and mortality worldwide, mainly in children, immunocompromised people, and the elderly. Several respiratory viruses can induce intestinal inflammation and alterations in intestinal microbiota composition. Human metapneumovirus (HMPV) is one of the major respiratory viruses contributing to infant mortality in children under 5 years of age worldwide, and the effect of this infection at the gut level has not been studied. Methods Here, we evaluated the distal effects of HMPV infection on intestinal microbiota and inflammation in a murine model, analyzing several post-infection times (days 1, 3, and 5). Six to eight-week-old C57BL/6 mice were infected intranasally with HMPV, and mice inoculated with a non-infectious supernatant (Mock) were used as a control group. Results We did not detect HMPV viral load in the intestine, but we observed significant changes in the transcription of IFN-γ in the colon, analyzed by qPCR, at day 1 post-infection as compared to the control group. Furthermore, we analyzed the frequencies of different innate and adaptive immune cells in the colonic lamina propria, using flow cytometry. The frequency of monocyte populations was altered in the colon of HMPV -infected mice at days 1 and 3, with no significant difference from control mice at day 5 post-infection. Moreover, colonic CD8+ T cells and memory precursor effector CD8+ T cells were significantly increased in HMPV-infected mice at day 5, suggesting that HMPV may also alter intestinal adaptive immunity. Additionally, we did not find alterations in antimicrobial peptide expression, the frequency of colonic IgA+ plasma cells, and levels of fecal IgA. Some minor alterations in the fecal microbiota composition of HMPV -infected mice were detected using 16s rRNA sequencing. However, no significant differences were found in β-diversity and relative abundance at the genus level. Discussion To our knowledge, this is the first report describing the alterations in intestinal immunity following respiratory infection with HMPV infection. These effects do not seem to be mediated by direct viral infection in the intestinal tract. Our results indicate that HMPV can affect colonic innate and adaptive immunity but does not significantly alter the microbiota composition, and further research is required to understand the mechanisms inducing these distal effects in the intestine.
Collapse
Affiliation(s)
- Javiera Sepúlveda-Alfaro
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eduardo A. Catalán
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Omar P. Vallejos
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio Ramos-Tapia
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | | | - María J. Mendoza-León
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Isidora D. Suazo
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Elizabeth Rivera-Asin
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Pedro H. Silva
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Oscar Alvarez-Mardones
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | | | - Claudia A. Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | | | - Juan A. Ugalde
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Jorge A. Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Melo-Gonzalez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| |
Collapse
|
17
|
Leleiwi I, Kokkinias K, Kim Y, Baniasad M, Shaffer M, Sabag-Daigle A, Daly RA, Flynn RM, Wysocki VH, Ahmer BMM, Borton MA, Wrighton KC. Gut microbiome carbon and sulfur metabolisms support Salmonella during pathogen infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.16.575907. [PMID: 38293109 PMCID: PMC10827160 DOI: 10.1101/2024.01.16.575907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Salmonella enterica serovar Typhimurium is a pervasive enteric pathogen and an ongoing global threat to public health. Ecological studies in the Salmonella impacted gut remain underrepresented in the literature, discounting the microbiome mediated interactions that may inform Salmonella physiology during colonization and infection. To understand the microbial ecology of Salmonella remodeling of the gut microbiome, here we performed multi-omics approaches on fecal microbial communities from untreated and Salmonella -infected mice. Reconstructed genomes recruited metatranscriptomic and metabolomic data providing a strain-resolved view of the expressed metabolisms of the microbiome during Salmonella infection. This data informed possible Salmonella interactions with members of the gut microbiome that were previously uncharacterized. Salmonella- induced inflammation significantly reduced the diversity of transcriptionally active members in the gut microbiome, yet increased gene expression was detected for 7 members, with Luxibacter and Ligilactobacillus being the most active. Metatranscriptomic insights from Salmonella and other persistent taxa in the inflamed microbiome further expounded the necessity for oxidative tolerance mechanisms to endure the host inflammatory responses to infection. In the inflamed gut lactate was a key metabolite, with microbiota production and consumption reported amongst transcriptionally active members. We also showed that organic sulfur sources could be converted by gut microbiota to yield inorganic sulfur pools that become oxidized in the inflamed gut, resulting in thiosulfate and tetrathionate that supports Salmonella respiration. Advancement of pathobiome understanding beyond inferences from prior amplicon-based approaches can hold promise for infection mitigation, with the active community outlined here offering intriguing organismal and metabolic therapeutic targets.
Collapse
|
18
|
Leleiwi I, Kokkinias K, Kim Y, Baniasad M, Shaffer M, Sabag-Daigle A, Daly RA, Flynn RM, Wysocki VH, Ahmer BMM, Borton MA, Wrighton KC. Gut microbiota carbon and sulfur metabolisms support Salmonella infections. THE ISME JOURNAL 2024; 18:wrae187. [PMID: 39404095 PMCID: PMC11482014 DOI: 10.1093/ismejo/wrae187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/15/2024] [Indexed: 10/18/2024]
Abstract
Salmonella enterica serovar Typhimurium is a pervasive enteric pathogen and ongoing global threat to public health. Ecological studies in the Salmonella impacted gut remain underrepresented in the literature, discounting microbiome mediated interactions that may inform Salmonella physiology during colonization and infection. To understand the microbial ecology of Salmonella remodeling of the gut microbiome, we performed multi-omics on fecal microbial communities from untreated and Salmonella-infected mice. Reconstructed genomes recruited metatranscriptomic and metabolomic data providing a strain-resolved view of the expressed metabolisms of the microbiome during Salmonella infection. These data informed possible Salmonella interactions with members of the gut microbiome that were previously uncharacterized. Salmonella-induced inflammation significantly reduced the diversity of genomes that recruited transcripts in the gut microbiome, yet increased transcript mapping was observed for seven members, among which Luxibacter and Ligilactobacillus transcript read recruitment was most prevalent. Metatranscriptomic insights from Salmonella and other persistent taxa in the inflamed microbiome further expounded the necessity for oxidative tolerance mechanisms to endure the host inflammatory responses to infection. In the inflamed gut lactate was a key metabolite, with microbiota production and consumption reported amongst members with detected transcript recruitment. We also showed that organic sulfur sources could be converted by gut microbiota to yield inorganic sulfur pools that become oxidized in the inflamed gut, resulting in thiosulfate and tetrathionate that support Salmonella respiration. This research advances physiological microbiome insights beyond prior amplicon-based approaches, with the transcriptionally active organismal and metabolic pathways outlined here offering intriguing intervention targets in the Salmonella-infected intestine.
Collapse
Affiliation(s)
- Ikaia Leleiwi
- Department of Cell and Molecular Biology, Colorado State University, Plant Sciences Bldg. 307 University Ave, Fort Collins, CO 80523, United States
- Department of Soil and Crop Sciences, Colorado State University, Plant Sciences Bldg. 307 University Ave, Fort Collins, CO 80523, United States
| | - Katherine Kokkinias
- Department of Soil and Crop Sciences, Colorado State University, Plant Sciences Bldg. 307 University Ave, Fort Collins, CO 80523, United States
- Department of Microbiology, Immunology, and Pathology, Microbiology Building, 1682 Campus Delivery Colorado State University, Fort Collins, CO 80523, United States
| | - Yongseok Kim
- Department of Chemistry and Biochemistry, The Ohio State University, 200 CBEC Building 151 W. Woodruff Ave. Columbus, OH 43210, United States
| | - Maryam Baniasad
- Department of Chemistry and Biochemistry, The Ohio State University, 200 CBEC Building 151 W. Woodruff Ave. Columbus, OH 43210, United States
| | - Michael Shaffer
- Department of Soil and Crop Sciences, Colorado State University, Plant Sciences Bldg. 307 University Ave, Fort Collins, CO 80523, United States
| | - Anice Sabag-Daigle
- Department of Microbial Infection and immunity, The Ohio State University, 776 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, OH 43210-2210, United States
| | - Rebecca A Daly
- Department of Soil and Crop Sciences, Colorado State University, Plant Sciences Bldg. 307 University Ave, Fort Collins, CO 80523, United States
| | - Rory M Flynn
- Department of Soil and Crop Sciences, Colorado State University, Plant Sciences Bldg. 307 University Ave, Fort Collins, CO 80523, United States
| | - Vicki H Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, 200 CBEC Building 151 W. Woodruff Ave. Columbus, OH 43210, United States
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, 280 Biomedical Research Tower 460 W. 12th Ave. Columbus, OH 43210, United States
| | - Brian M M Ahmer
- Department of Microbial Infection and immunity, The Ohio State University, 776 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, OH 43210-2210, United States
| | - Mikayla A Borton
- Department of Soil and Crop Sciences, Colorado State University, Plant Sciences Bldg. 307 University Ave, Fort Collins, CO 80523, United States
| | - Kelly C Wrighton
- Department of Cell and Molecular Biology, Colorado State University, Plant Sciences Bldg. 307 University Ave, Fort Collins, CO 80523, United States
- Department of Soil and Crop Sciences, Colorado State University, Plant Sciences Bldg. 307 University Ave, Fort Collins, CO 80523, United States
- Department of Microbiology, Immunology, and Pathology, Microbiology Building, 1682 Campus Delivery Colorado State University, Fort Collins, CO 80523, United States
| |
Collapse
|
19
|
Yonoichi S, Hirano T, Hara Y, Ishida Y, Shoda A, Kimura M, Murata M, Mantani Y, Yokoyama T, Ikenaka Y, Hoshi N. Effects of exposure to the neonicotinoid pesticide clothianidin on mouse intestinal microbiota under unpredictable environmental stress. Toxicol Appl Pharmacol 2024; 482:116795. [PMID: 38160895 DOI: 10.1016/j.taap.2023.116795] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/28/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
Recent research has demonstrated the toxicity of neonicotinoid pesticides (NNs) in mammals through their interaction with nicotinic acetylcholine receptors (nAChRs). These effects are reported to extend to the intestinal microbiota as well. In addition, environmental stress affects the expression of nAChRs, which may alter sensitivity to NNs. In this study, we analyzed the intestinal microbiota of mice exposed to clothianidin (CLO), a type of NN, under environmental stress, and aimed to clarify the effects of such combined exposure on the intestinal microbiota. C57BL/6N male mice (9 weeks old) were subchronically administered a no-observed-adverse-effect-level (NOAEL) CLO-mixed rehydration gel for 29 days and simultaneously subjected to chronic unpredictable mild stress (CUMS). After the administration period, cecum contents were collected and analyzed by 16S rRNA sequencing for intestinal microbiota. CLO exposure alone resulted in alterations in the relative abundance of Alistipes and ASF356, which produce short-chain fatty acids. The addition of CUMS amplified these changes. On the other hand, CLO alone did not affect the relative abundance of Lactobacillus, but the abundance decreased when CUMS was added. This study revealed that the combined exposure to CLO and stress not only amplifies their individual effects on intestinal microbiota but also demonstrates combined and multifaceted toxicities.
Collapse
Affiliation(s)
- Sakura Yonoichi
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Tetsushi Hirano
- Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Yukako Hara
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Yuya Ishida
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Asuka Shoda
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Mako Kimura
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Midori Murata
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Youhei Mantani
- Laboratory of Histophysiology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Toshifumi Yokoyama
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Yoshinori Ikenaka
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan; Translational Research Unit, Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan; One Health Research Center, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan; Water Research Group, Unit for Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| | - Nobuhiko Hoshi
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan.
| |
Collapse
|
20
|
Zhang W, Dong XY, Huang R. Gut Microbiota in Ischemic Stroke: Role of Gut Bacteria-Derived Metabolites. Transl Stroke Res 2023; 14:811-828. [PMID: 36279071 DOI: 10.1007/s12975-022-01096-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/05/2022] [Accepted: 10/07/2022] [Indexed: 11/30/2022]
Abstract
Ischemic stroke (IS) remains a leading cause of death and long-term disability globally. Several mechanisms including glutamate excitotoxicity, calcium overload, neuroinflammation, oxidative stress, mitochondrial damage, and apoptosis are known to be involved in the pathogenesis of IS, but the underlying pathophysiology mechanisms of IS are not fully clarified. During the past decade, gut microbiota were recognized as a key regulator to affect the health of the host either directly or via their metabolites. Recent studies indicate that gut bacterial dysbiosis is closely related to hypertension, diabetes, obesity, dyslipidemia, and metabolic syndrome, which are the main risk factors for cardiovascular diseases. Increasing evidence indicates that IS can lead to perturbation in gut microbiota and increased permeability of the gut mucosa, known as "leaky gut," resulting in endotoxemia and bacterial translocation. In turn, gut dysbiosis and impaired intestinal permeability can alter gut bacterial metabolite signaling profile from the gut to the brain. Microbiota-derived products and metabolites, such as short-chain fatty acids (SCFAs), bile acids (BAs), trimethylamine N-oxide (TMAO), lipopolysaccharides (LPS), and phenylacetylglutamine (PAGln) can exert beneficial or detrimental effects on various extraintestinal organs, including the brain, liver, and heart. These metabolites have been increasingly acknowledged as biomarkers and mediators of IS. However, the specific role of the gut bacterial metabolites in the context of stroke remains incompletely understood. In-depth studies on these products and metabolites may provide new insight for the development of novel therapeutics for IS.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiao Yu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Huang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
21
|
Ren Y, Wang Y, Chen J, Fu S, Bu W, Xue H. Integrated analysis of miRNA profiles and gut bacterial changes in Altica viridicyanea following antibiotic treatment. Ecol Evol 2023; 13:e10660. [PMID: 37915809 PMCID: PMC10616750 DOI: 10.1002/ece3.10660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/29/2023] [Accepted: 10/12/2023] [Indexed: 11/03/2023] Open
Abstract
The gut bacteria involves in insect homeostasis by playing essential roles in host physiology, metabolism, innate immunity, and so forth. microRNAs (miRNAs) are endogenous small noncoding RNAs that posttranscriptionally regulate gene expression to affect immune or metabolic processes in insects. For several non-model insects, the available knowledge on the relationship between changes in the gut bacteria and miRNA profiles is limited. In this study, we investigated the gut bacterial diversity, composition, and function from Altica viridicyanea feeding on normal- and antibiotic-treated host plants using 16S rRNA amplicon sequencing; antibiotics have been shown to affect the body weight and development time in A. viridicyanea, suggesting that the gut bacteria of the normal sample were more diverse and abundant than those of the antibiotic-fed group, and most of them were involved in various physical functions by enrichment analysis. Furthermore, we executed small RNA transcriptome sequencing using the two experimental groups to obtain numerous sRNAs, such as piRNAs, siRNAs, and known and novel miRNAs, by data mapping and quality control, and furthermore, a total of 224 miRNAs were identified as significantly differentially expressed miRNAs, of which some DEMs and their target genes participated in immune- and metabolism-related pathways based on GO and KEGG annotation. Besides, regarding the regulatory roles of miRNA and target genes, a interaction network of DEM-target gene pairs from eight immune- or metabolism-related signaling pathways were constructed. Finally, we discovered that DEMs from above pathways were significantly positively or negatively correlated with gut bacterial alterations following antibiotic treatment. Collectively, the observations of this study expand our understanding of how the disturbance of gut bacteria affects miRNA profiles in A. viridicyanea and provide new valuable resources from extreme ranges for future studies on the adaptive evolution in insects.
Collapse
Affiliation(s)
- Yipeng Ren
- Institute of Entomology, College of Life SciencesNankai UniversityTianjinChina
| | - Yuan Wang
- Institute of Entomology, College of Life SciencesNankai UniversityTianjinChina
| | - Juhong Chen
- Institute of Entomology, College of Life SciencesNankai UniversityTianjinChina
| | - Siying Fu
- Institute of Entomology, College of Life SciencesNankai UniversityTianjinChina
| | - Wenjun Bu
- Institute of Entomology, College of Life SciencesNankai UniversityTianjinChina
| | - Huaijun Xue
- Institute of Entomology, College of Life SciencesNankai UniversityTianjinChina
| |
Collapse
|
22
|
Xu N, Zhang W, Huo J, Tao R, Jin T, Zhang Y, Wang Y, Zhu L, Li J, Yao Q, Ge L. Characterization of changes in the intestinal microbiome following combination therapy with zinc preparation and conventional treatment for children with rotavirus enteritis. Front Cell Infect Microbiol 2023; 13:1153701. [PMID: 37842003 PMCID: PMC10570505 DOI: 10.3389/fcimb.2023.1153701] [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: 01/30/2023] [Accepted: 09/06/2023] [Indexed: 10/17/2023] Open
Abstract
Background Rotavirus (RV) is one of the most common pathogens causing diarrhea in infants and young children worldwide. Routinely, antiviral therapy, intestinal mucosa protection, and fluid supplementation are used in clinic, however this is not efficacious in some severe cases. Zinc supplementation has previously been shown to improve resolution of symptoms from infectious diarrhea. Methods In this study differences in response rate, duration of hyperthermia, vomiting, and diarrhea, and the persistence time of cough and lung rales in groups were compared. 16SrDNA gene sequencing technology was used to analyze and compare changes in the intestinal microflora of children with RV enteritis who received the conventional treatment with or without the zinc preparation. In addition, the correlations between the differential bacterial species and the related inflammatory factors were determined. Results Conventional therapy combined with the zinc preparation significantly shortened the duration of hyperthermia, vomiting, and diarrhea compared with the conventional treatment alone. In addition, the time to symptom relief showed that the absorption time of cough and lung rales was significantly shorter in the combination treatment group than that in the conventional treatment group in the children with pneumonia. Further, compared with the conventional treatment, the combined treatment significantly increased the diversity and abundances of florae as compared with the conventional treatment. This combination therapy containing zinc preparation markedly increased the abundances of Faecalibacterium, Bacteroidales, Ruminoccoccoccus, and Lachnospiraceae at the genus level. The LEfSe analysis suggested that Clostridiumbolteae were most significantly altered after the combination therapy. In addition, a correlation analysis revealed significantly negative correlations between the inflammatory factors especially IL-6, TNF-a, CRP and some intestinal florae such as Bacteroides, Faecalibacterium, Blautia, Parabacteroides, Subdoligranulum, and Flavonifractor. Conclusion Compared with the conventional therapy alone, the combined therapy with the zinc preparation significantly improves symptoms caused by RV. The combination therapy containing the zinc preparation significantly increases the diversity and abundances of some beneficial groups of bacteria. Further, The presence of these groups was further negatively correlated with relevant inflammatory factors. More importantly, this combination therapy containing the zinc preparation provides a reference for the clinical management of children with RV enteritis.
Collapse
Affiliation(s)
- Ning Xu
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Wen Zhang
- Department of Gastroenterology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Jingjing Huo
- Department of Reproductive Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Rui Tao
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Taicheng Jin
- Department of Government, Hamilton College, Clinton, UT, United States
| | - Yuanmou Zhang
- Department of Gastroenterology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Yanjiao Wang
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Lei Zhu
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - JiaJia Li
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Qi Yao
- Department of Pharmacy, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Li Ge
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
23
|
Rui J, Zhao Y, Cong N, Wang F, Li C, Liu X, Hu J, Ling N, Jing X. Elevational distribution and seasonal dynamics of alpine soil prokaryotic communities. Front Microbiol 2023; 14:1280011. [PMID: 37808282 PMCID: PMC10557256 DOI: 10.3389/fmicb.2023.1280011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
The alpine grassland ecosystem is a biodiversity hotspot of plants on the Qinghai-Tibetan Plateau, where rapid climate change is altering the patterns of plant biodiversity along elevational and seasonal gradients of environments. However, how belowground microbial biodiversity changes along elevational gradient during the growing season is not well understood yet. Here, we investigated the elevational distribution of soil prokaryotic communities by using 16S rRNA amplicon sequencing along an elevational gradient between 3,200 and 4,200 m, and a seasonal gradient between June and September in the Qinghai-Tibetan alpine grasslands. First, we found soil prokaryotic diversity and community composition significantly shifted along the elevational gradient, mainly driven by soil temperature and moisture. Species richness did not show consistent elevational trends, while those of evenness declined with elevation. Copiotrophs and symbiotic diazotrophs declined with elevation, while oligotrophs and AOB increased, affected by temperature. Anaerobic or facultatively anaerobic bacteria and AOA were hump-shaped, mainly influenced by moisture. Second, seasonal patterns of community composition were mainly driven by aboveground biomass, precipitation, and soil temperature. The seasonal dynamics of community composition indicated that soil prokaryotic community, particularly Actinobacteria, was sensitive to short-term climate change, such as the monthly precipitation variation. At last, dispersal limitation consistently dominated the assembly process of soil prokaryotic communities along both elevational and seasonal gradients, especially for those of rare species, while the deterministic process of abundant species was relatively higher at drier sites and in drier July. The balance between deterministic and stochastic processes in abundant subcommunities might be strongly influenced by water conditions (precipitation/moisture). Our findings suggest that both elevation and season can alter the patterns of soil prokaryotic biodiversity in alpine grassland ecosystem of Qinghai-Tibetan Plateau, which is a biodiversity hotspot and is experiencing rapid climate change. This work provides new insights into the response of soil prokaryotic communities to changes in elevation and season, and helps us understand the temporal and spatial variations in such climate change-sensitive regions.
Collapse
Affiliation(s)
- Junpeng Rui
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
- Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Yuwei Zhao
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
- Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Nan Cong
- Lhasa Plateau Ecosystem Research Station, Institute of Geographic Sciences and Natural Resources Research, Chinese Academy of Sciences, Beijing, China
| | - Fuxin Wang
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Chao Li
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Xiang Liu
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Jingjing Hu
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Ning Ling
- Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Xin Jing
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| |
Collapse
|
24
|
Gao Y, Wu X, Zhao N, Bai D. Scientific connotation of the compatibility of traditional Chinese medicine from the perspective of the intestinal flora. Front Pharmacol 2023; 14:1152858. [PMID: 37538183 PMCID: PMC10395102 DOI: 10.3389/fphar.2023.1152858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 06/26/2023] [Indexed: 08/05/2023] Open
Abstract
Revealing the connotation of the compatibility of Chinese medicines (CM) is a requirement for the modernization of traditional Chinese medicine (TCM). However, no consensus exists on the specific mechanism of traditional Chinese medicine compatibility (TCMC). Many studies have shown that the occurrence and development of diseases and the efficacy of CM are closely related to intestinal flora (IF), which may provide a new perspective to understand the theory of TCM. This study aimed to summarize the relationship between the changes in IF before and after the compatibility of different drugs and the synergistic, toxicity reduction, and incompatibility effects of drug pairs from the perspective of the effects of CM on the IF and the regulation of microbial metabolites. These studies showed that the effect of drug pairs on the composition of the IF is not a simple superposition of two single drugs, and that the drug pairs also play a specific role in regulating the production of intestinal bacterial metabolites; therefore, it has a different pharmacodynamic effect, which may provide a perspective to clarify the compatibility mechanism. However, research on the interpretation of the scientific connotations of TCMC from the perspective of the IF is still in its infancy and has limitations. Therefore, this study aimed to summarize previous research experience and proposed to conduct a deep and systematic study from the perspective of drug pair dismantling, IF, intestinal bacteria metabolite, organism, and disease to provide a reference for scientific research on the compatibility mechanism of CM.
Collapse
Affiliation(s)
- Yuan Gao
- Fang Zheng Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxia Wu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ning Zhao
- Fang Zheng Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Pharmacy, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dong Bai
- Fang Zheng Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Jiang F, Song P, Liu D, Zhang J, Qin W, Wang H, Liang C, Gao H, Zhang T. Marked variations in gut microbial diversity, functions, and disease risk between wild and captive alpine musk deer. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12675-1. [PMID: 37421471 PMCID: PMC10390370 DOI: 10.1007/s00253-023-12675-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 07/10/2023]
Abstract
Maintaining a healthy status is crucial for the successful captive breeding of endangered alpine musk deer (Moschus chrysogaster, AMD), and captive breeding programs are beneficial to the ex-situ conservation and wild population recovery of this species. Meanwhile, the gut microbiota is essential for host health, survival, and environmental adaptation. However, changes in feeding environment and food can affect the composition and function of gut microbiota in musk deer, ultimately impacting their health and adaptation. Therefore, regulating the health status of wild and captive AMD through a non-invasive method that targets gut microbiota is a promising approach. Here, 16S rRNA gene sequencing was employed to reveal the composition and functional variations between wild (N = 23) and captive (N = 25) AMD populations. The results indicated that the gut microbiota of wild AMD exhibited significantly higher alpha diversity (P < 0.001) and greater abundance of the phylum Firmicutes, as well as several dominant genera, including UCG-005, Christensenellaceae R7 group, Monoglobus, Ruminococcus, and Roseburia (P < 0.05), compared to captive AMD. These findings suggest that the wild AMD may possess more effective nutrient absorption and utilization, a more stable intestinal microecology, and better adaption to the complex natural environment. The captive individuals displayed higher metabolic functions with an increased abundance of the phylum Bacteroidetes and certain dominant genera, including Bacteroides, Rikenellaceae RC9 gut group, NK4A214 group, and Alistipes (P < 0.05), which contributed to the metabolic activities of various nutrients. Furthermore, captive AMD showed a higher level of 11 potential opportunistic pathogens and a greater enrichment of disease-related functions compared to wild AMD, indicating that wild musk deer have a lower risk of intestinal diseases and more stable intestinal structure in comparison to captive populations. These findings can serve as a valuable theoretical foundation for promoting the healthy breeding of musk deer and as a guide for evaluating the health of wild-released and reintroduced musk deer in the future. KEY POINTS: • Wild and captive AMD exhibit contrasting gut microbial diversity and certain functions. • With higher diversity, certain bacteria aid wild AMD's adaptation to complex habitats. • Higher potential pathogens and functions increase disease risk in captive AMD.
Collapse
Affiliation(s)
- Feng Jiang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, 23 Xinning Rd, Chengxi District, Qinghai, 810001, Xining, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, 810001, Qinghai, China
| | - Pengfei Song
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, 23 Xinning Rd, Chengxi District, Qinghai, 810001, Xining, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, 810001, Qinghai, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Daoxin Liu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, 23 Xinning Rd, Chengxi District, Qinghai, 810001, Xining, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, 810001, Qinghai, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jingjie Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, 23 Xinning Rd, Chengxi District, Qinghai, 810001, Xining, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, 810001, Qinghai, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wen Qin
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, 810016, Qinghai, China
| | - Haijing Wang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, 23 Xinning Rd, Chengxi District, Qinghai, 810001, Xining, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, 810001, Qinghai, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chengbo Liang
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, 810016, Qinghai, China
| | - Hongmei Gao
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, 23 Xinning Rd, Chengxi District, Qinghai, 810001, Xining, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, 810001, Qinghai, China
| | - Tongzuo Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, 23 Xinning Rd, Chengxi District, Qinghai, 810001, Xining, China.
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, 810001, Qinghai, China.
| |
Collapse
|
26
|
Lv H, Jia H, Cai W, Cao R, Xue C, Dong N. Rehmannia glutinosa polysaccharides attenuates colitis via reshaping gut microbiota and short-chain fatty acid production. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:3926-3938. [PMID: 36347632 DOI: 10.1002/jsfa.12326] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/23/2022] [Accepted: 11/09/2022] [Indexed: 05/03/2023]
Abstract
BACKGROUND Ulcerative colitis is a gastrointestinal disease closely related to intestinal epithelial barrier damage and intestinal microbiome imbalance; however, effective treatment methods are currently limited. Rehmannia glutinosa polysaccharide (RGP) is an important active ingredient with a wide range of pharmacological activities, although its protective effect on colitis remains to be explored. In the present study, we verified the in vitro anti-inflammatory effect of RGP, and observed the ameliorating effect of RGP on dextran sulfate sodium-induced colitis in mice. RESULTS The results showed that (i) RGP attenuates lipopolysaccharide-induced overexpression of inflammatory factors in RAW264.7 cells; (ii) RGP improves the pathological damage caused by DSS, including weight loss, increased disease activity index and intestinal tissue ulcers; (iii) RGP improves tight junction proteins to protects the tightness of the intestinal epithelium; (iv) RGP inhibits the expression of inflammatory factors through the nuclear factor-kappa B pathway, and improved the of intestinal tissues inflammation; and (v) RGP can maintain the species diversity of intestinal microbes, increase the content of short-chain fatty acids and then restore the imbalance of intestinal microecology. CONCLUSION RGP can improve the intestinal microbiota to strengthen the intestinal epithelial barrier and protect against DSS-induced colitis. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Hao Lv
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Hongpeng Jia
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Wenjie Cai
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Rujing Cao
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Chenyu Xue
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Na Dong
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
27
|
Leleiwi I, Rodriguez-Ramos J, Shaffer M, Sabag-Daigle A, Kokkinias K, Flynn RM, Daly RA, Kop LFM, Solden LM, Ahmer BMM, Borton MA, Wrighton KC. Exposing new taxonomic variation with inflammation - a murine model-specific genome database for gut microbiome researchers. MICROBIOME 2023; 11:114. [PMID: 37210515 PMCID: PMC10199544 DOI: 10.1186/s40168-023-01529-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/21/2023] [Indexed: 05/22/2023]
Abstract
BACKGROUND The murine CBA/J mouse model widely supports immunology and enteric pathogen research. This model has illuminated Salmonella interactions with the gut microbiome since pathogen proliferation does not require disruptive pretreatment of the native microbiota, nor does it become systemic, thereby representing an analog to gastroenteritis disease progression in humans. Despite the value to broad research communities, microbiota in CBA/J mice are not represented in current murine microbiome genome catalogs. RESULTS Here we present the first microbial and viral genomic catalog of the CBA/J murine gut microbiome. Using fecal microbial communities from untreated and Salmonella-infected, highly inflamed mice, we performed genomic reconstruction to determine the impacts on gut microbiome membership and functional potential. From high depth whole community sequencing (~ 42.4 Gbps/sample), we reconstructed 2281 bacterial and 4516 viral draft genomes. Salmonella challenge significantly altered gut membership in CBA/J mice, revealing 30 genera and 98 species that were conditionally rare and unsampled in non-inflamed mice. Additionally, inflamed communities were depleted in microbial genes that modulate host anti-inflammatory pathways and enriched in genes for respiratory energy generation. Our findings suggest decreases in butyrate concentrations during Salmonella infection corresponded to reductions in the relative abundance in members of the Alistipes. Strain-level comparison of CBA/J microbial genomes to prominent murine gut microbiome databases identified newly sampled lineages in this resource, while comparisons to human gut microbiomes extended the host relevance of dominant CBA/J inflammation-resistant strains. CONCLUSIONS This CBA/J microbiome database provides the first genomic sampling of relevant, uncultivated microorganisms within the gut from this widely used laboratory model. Using this resource, we curated a functional, strain-resolved view on how Salmonella remodels intact murine gut communities, advancing pathobiome understanding beyond inferences from prior amplicon-based approaches. Salmonella-induced inflammation suppressed Alistipes and other dominant members, while rarer commensals like Lactobacillus and Enterococcus endure. The rare and novel species sampled across this inflammation gradient advance the utility of this microbiome resource to benefit the broad research needs of the CBA/J scientific community, and those using murine models for understanding the impact of inflammation on the gut microbiome more generally. Video Abstract.
Collapse
Affiliation(s)
- Ikaia Leleiwi
- Department of Cell and Molecular Biology, The Colorado State University, Fort Collins, CO USA
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO USA
| | - Josué Rodriguez-Ramos
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO USA
- Graduate Degree Program in Ecology, The Colorado State University, Fort Collins, CO USA
| | - Michael Shaffer
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO USA
| | - Anice Sabag-Daigle
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH USA
| | - Katherine Kokkinias
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO USA
- Department of Microbiology, Immunology, and Pathology, The Colorado State University, Fort Collins, CO USA
| | - Rory M. Flynn
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO USA
| | - Rebecca A. Daly
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO USA
| | - Linnea F. M. Kop
- Department of Microbiology, RIBES, Radbound University, Nijmegen, The Netherlands
- Department of Microbiology and Biophysics, The Ohio State University, Columbus, OH USA
| | - Lindsey M. Solden
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO USA
| | - Brian M. M. Ahmer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH USA
| | - Mikayla A. Borton
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO USA
| | - Kelly C. Wrighton
- Department of Cell and Molecular Biology, The Colorado State University, Fort Collins, CO USA
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO USA
- Graduate Degree Program in Ecology, The Colorado State University, Fort Collins, CO USA
- Department of Microbiology, Immunology, and Pathology, The Colorado State University, Fort Collins, CO USA
| |
Collapse
|
28
|
Chen L, Guo L, Feng S, Wang C, Cui Z, Wang S, Lu Q, Chang H, Hang B, Snijders AM, Mao JH, Lu Y, Ding D. Fecal microbiota transplantation ameliorates type 2 diabetes via metabolic remodeling of the gut microbiota in db/db mice. BMJ Open Diabetes Res Care 2023; 11:e003282. [PMID: 37253485 PMCID: PMC10230930 DOI: 10.1136/bmjdrc-2022-003282] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 04/30/2023] [Indexed: 06/01/2023] Open
Abstract
INTRODUCTION Gut microbiome (GM) deregulation has been implicated in major conditions such as obesity and type 2 diabetes (T2DM). Our previous prospective study indicated that fecal microbiota transplantation (FMT) successfully improved patients with T2DM. We hypothesized that FMT may be a potential therapeutic method for T2DM, but its precise mechanisms in T2DM remains to be elucidated. RESEARCH DESIGN AND METHODS Eight db/m mice were FMT donors and control mice, and 16 genetically diabetic db/db mice were equally divided into two groups (db/db+phosphate-buffered saline (PBS) group, db/db+FMT group). The db/db+FMT group was administered fresh fecal suspension (0.2 mL/mice) daily for 4 weeks. Analysis of the GM and serum metabolome was carried out by 16S ribosomal RNA sequencing and liquid chromatogram-mass spectrometry, respectively. Effects of FMT on the gut barrier and pancreas were assessed using protein assays, messenger RNA, immunohistology and clinical indicators testing. RESULTS Our results showed that FMT treatment of db/db mice relieves a series of clinical indicators, including fasting plasma glucose, serum insulin and oral glucose tolerance test among others. Compared with non-diabetic control mice, db/db+PBS mice exhibited decreased abundance of Ruminococaceae, Porphyromonadaceae and increased abundance of Rikenellaceae and Lactobacillaceae. FMT treatment reversed this effect on the microbiome. Eleven metabolites were changed between the db/db+PBS and db/db+FMT groups. Correlation analysis showed that the structural changes of the GM were correlated with host metabolite levels. We further showed that FMT treatment of db/db mice improved intestinal barrier function, reduced inflammation and caused an alteration in the number of circulating immune cells. CONCLUSIONS FMT-mediated changes in the GM, serum metabolites, intestinal epithelial barrier, inflammation and circulating immune cells play an important role in the efficacy of FMT on T2DM disease progression.
Collapse
Affiliation(s)
- Lijuan Chen
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University Second Affiliated Hospital, Nanjing, Jiangsu, China
| | - Lin Guo
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University Second Affiliated Hospital, Nanjing, Jiangsu, China
| | - Susu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University Second Affiliated Hospital, Nanjing, Jiangsu, China
| | - Congcong Wang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University Second Affiliated Hospital, Nanjing, Jiangsu, China
| | - Zhicheng Cui
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University Second Affiliated Hospital, Nanjing, Jiangsu, China
| | - Sijing Wang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University Second Affiliated Hospital, Nanjing, Jiangsu, China
| | - Qingmiao Lu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University Second Affiliated Hospital, Nanjing, Jiangsu, China
| | - Hang Chang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Bo Hang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
- Department of Biomaterials, Berkeley-Nanjing Research Center, Nanjing, Jiangsu, China
| | - Antoine M Snijders
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Yibing Lu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University Second Affiliated Hospital, Nanjing, Jiangsu, China
| | - Dafa Ding
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University Second Affiliated Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
29
|
Mazumder MHH, Gandhi J, Majumder N, Wang L, Cumming RI, Stradtman S, Velayutham M, Hathaway QA, Shannahan J, Hu G, Nurkiewicz TR, Tighe RM, Kelley EE, Hussain S. Lung-gut axis of microbiome alterations following co-exposure to ultrafine carbon black and ozone. Part Fibre Toxicol 2023; 20:15. [PMID: 37085867 PMCID: PMC10122302 DOI: 10.1186/s12989-023-00528-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Microbial dysbiosis is a potential mediator of air pollution-induced adverse outcomes. However, a systemic comparison of the lung and gut microbiome alterations and lung-gut axis following air pollution exposure is scant. In this study, we exposed male C57BL/6J mice to inhaled air, CB (10 mg/m3), O3 (2 ppm) or CB + O3 mixture for 3 h/day for either one day or four consecutive days and were euthanized 24 h post last exposure. The lung and gut microbiome were quantified by 16 s sequencing. RESULTS Multiple CB + O3 exposures induced an increase in the lung inflammatory cells (neutrophils, eosinophils and B lymphocytes), reduced absolute bacterial load in the lungs and increased load in the gut. CB + O3 exposure was more potent as it decreased lung microbiome alpha diversity just after a single exposure. CB + O3 co-exposure uniquely increased Clostridiaceae and Prevotellaceae in the lungs. Serum short chain fatty acids (SCFA) (acetate and propionate) were increased significantly only after CB + O3 co-exposure. A significant increase in SCFA producing bacterial families (Ruminococcaceae, Lachnospiraceae, and Eubacterium) were also observed in the gut after multiple exposures. Co-exposure induced significant alterations in the gut derived metabolite receptors/mediator (Gcg, Glp-1r, Cck) mRNA expression. Oxidative stress related mRNA expression in lungs, and oxidant levels in the BALF, serum and gut significantly increased after CB + O3 exposures. CONCLUSION Our study confirms distinct gut and lung microbiome alterations after CB + O3 inhalation co-exposure and indicate a potential homeostatic shift in the gut microbiome to counter deleterious impacts of environmental exposures on metabolic system.
Collapse
Affiliation(s)
- Md Habibul Hasan Mazumder
- Department of Physiology, Pharmacology, and Toxicology, Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Jasleen Gandhi
- Department of Microbiology, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Nairrita Majumder
- Department of Physiology, Pharmacology, and Toxicology, Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Lei Wang
- Department of Microbiology, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Robert Ian Cumming
- Department of Medicine, Duke University Medical Center, Durham, NC, 2927, USA
| | - Sydney Stradtman
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Murugesan Velayutham
- Department of Physiology, Pharmacology, and Toxicology, Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Quincy A Hathaway
- Heart and Vascular Institute, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Jonathan Shannahan
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Gangqing Hu
- Department of Microbiology, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Timothy R Nurkiewicz
- Department of Physiology, Pharmacology, and Toxicology, Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Robert M Tighe
- Department of Medicine, Duke University Medical Center, Durham, NC, 2927, USA
| | - Eric E Kelley
- Department of Physiology, Pharmacology, and Toxicology, Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Salik Hussain
- Department of Physiology, Pharmacology, and Toxicology, Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA.
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA.
- Department of Microbiology, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
30
|
Zhang K, Zhu L, Zhong Y, Xu L, Lang C, Chen J, Yan F, Li J, Qiu J, Chen Y, Sun D, Wang G, Qu K, Qin X, Wu W. Prodrug Integrated Envelope on Probiotics to Enhance Target Therapy for Ulcerative Colitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205422. [PMID: 36507607 PMCID: PMC9896077 DOI: 10.1002/advs.202205422] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/23/2022] [Indexed: 05/25/2023]
Abstract
Ulcerative colitis (UC), affecting millions of patients worldwide, is associated with disorders of the gut microbiota. Probiotics-based therapy positively regulating the community structure of gut microbiota is regarded as an efficient intervention for UC. However, oral probiotics delivery is restricted by limited bioactivity, short retention time, complex pathological condition, and single therapeutic efficacy. Here, a bioengineered probiotic decorated with a multifunctional prodrug coating is constructed to ameliorate the aforementioned shortcomings. The results of UC mice induced by dextran sulfate sodium demonstrate that the intrinsic features of the fabricated coating integrate gut microbes protection, colon-targeted drug release, prolonged drug retention, and inflammation regulation. In parallel, the probiotics Lactobacillus rhamnosus GG (LGG) could regulate the composition of the gut microbiota and improve epithelial barrier function, thereby synergistically ameliorating UC. These results provide ample shreds of evidence of the therapeutic effect on UC, therefore, demonstrate a great promise as the potential therapeutic strategy for UC treatment.
Collapse
Affiliation(s)
- Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030P. R. China
- Chongqing University Three Gorges HospitalChongqing Municipality Clinical Research Center for Geriatric diseasesChongqing404000P. R. China
| | - Li Zhu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030P. R. China
| | - Yuan Zhong
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030P. R. China
| | - Lixin Xu
- Chongqing University Three Gorges HospitalChongqing Municipality Clinical Research Center for Geriatric diseasesChongqing404000P. R. China
| | - Chunhui Lang
- Chongqing University Three Gorges HospitalChongqing Municipality Clinical Research Center for Geriatric diseasesChongqing404000P. R. China
| | - Jian Chen
- Chongqing University Three Gorges HospitalChongqing Municipality Clinical Research Center for Geriatric diseasesChongqing404000P. R. China
| | - Fei Yan
- Chongqing University Three Gorges HospitalChongqing Municipality Clinical Research Center for Geriatric diseasesChongqing404000P. R. China
| | - Jiawei Li
- Chongqing University Three Gorges HospitalChongqing Municipality Clinical Research Center for Geriatric diseasesChongqing404000P. R. China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030P. R. China
| | - Yidan Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030P. R. China
| | - Da Sun
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang ProvinceWenzhou UniversityWenzhouZhejiang325035P. R. China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030P. R. China
- Jin Feng LaboratoryChongqing401329P. R. China
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030P. R. China
- Chongqing University Three Gorges HospitalChongqing Municipality Clinical Research Center for Geriatric diseasesChongqing404000P. R. China
| | - Xian Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030P. R. China
- Chongqing University Three Gorges HospitalChongqing Municipality Clinical Research Center for Geriatric diseasesChongqing404000P. R. China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030P. R. China
- Jin Feng LaboratoryChongqing401329P. R. China
| |
Collapse
|
31
|
Paden H, Kurbatfinski N, Poelstra JW, Ormiston K, Orchard T, Ilic S. Dietary Impacts on Changes in Diversity and Abundance of the Murine Microbiome during Progression and Treatment of Cancer. Nutrients 2023; 15:724. [PMID: 36771430 PMCID: PMC9920799 DOI: 10.3390/nu15030724] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/14/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
The intestinal microbial population is recognized for its impact on cancer treatment outcomes. Little research has reported microbiome changes during cancer progression or the interplay of disease progression, dietary sugar/fat intake, and the microbiome through surgery and chemotherapy. In this study, the murine gut microbiome was used as a model system, and changes in microbiome diversity, richness, and evenness over the progression of the cancer and treatment were analyzed. Mice were categorized into four diet cohorts, combinations of either high or low sucrose and high or low omega-3 fatty acids, and two treatment cohorts, saline vehicle or chemotherapy, for a total of eight groups. Fecal samples were collected at specific timepoints to assess changes due to diet implementation, onset of cancer, lumpectomy, and chemotherapy. Akkermansia muciniphila abundance was very high in some samples and negatively correlated with overall Amplicon Sequence Variant (ASV) richness (r(64) = -0.55, p = 3 × 10-8). Throughout the disease progression, ASV richness significantly decreased and was impacted by diet and treatment. Alpha-diversity and differential microbial abundance were significantly affected by disease progression, diet, treatment, and their interactions. These findings help establish a baseline for understanding how cancer progression, dietary macronutrients, and specific treatments impact the murine microbiome, which may influence outcomes.
Collapse
Affiliation(s)
- Holly Paden
- Department of Human Sciences, Ohio State University, Columbus, OH 43210, USA
| | - Nikola Kurbatfinski
- Department of Human Sciences, Ohio State University, Columbus, OH 43210, USA
| | - Jelmer W. Poelstra
- Molecular and Cellular Imaging Center (MCIC), Ohio State University, Wooster, OH 44691, USA
| | - Kate Ormiston
- Department of Human Sciences, Ohio State University, Columbus, OH 43210, USA
| | - Tonya Orchard
- Department of Human Sciences, Ohio State University, Columbus, OH 43210, USA
| | - Sanja Ilic
- Department of Human Sciences, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
32
|
Gómez-Baltazar A, Vázquez-Marrufo G, Astiazaran-Garcia H, Ochoa-Zarzosa A, Canett-Romero R, García-Galaz A, Torres-Vega C, Vázquez-Garcidueñas MS. Comparative virulence of the worldwide ST19 and emergent ST213 genotypes of Salmonella enterica serotype Typhimurium strains isolated from food. Microbes Infect 2023; 25:105019. [PMID: 35781097 DOI: 10.1016/j.micinf.2022.105019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 02/04/2023]
Abstract
Salmonella enterica Typhimurium represents one of the most frequent causal agents of food contamination associated to gastroenteritis. The sequence type ST19 is the founder and worldwide prevalent genotype within this serotype, but its replacement by emerging genotypes has been recently reported. Particularly, the ST213 genotype has replaced it as the most prevalent in clinical and contaminated food samples in Mexico and has been recently reported in several countries. In this study, the in vitro and in vivo virulence of ST213 and ST19 strains isolated from food samples in Mexico was evaluated. Three out of the five analyzed ST213 strains, showed a greater internalization capacity and increased secretion of interleukins IL-8 and IL-6 of Caco-2 cells than the ST19 strains. Microbiological counts in feces and tissues showed the ability of all strains tested to establish infection in the rat model. The ST213 strains also caused histopathological damage, characteristic of gastroenteritis in Wistar rats. In contrast to the in vitro result, one of the ST19 strains showed marked damage in the test animals. The ST213 genotype strains showed in vitro and in vivo virulence variability, but significantly higher than the observed in the ST19 genotype strains, thus such emergent genotype represents a public health concern.
Collapse
Affiliation(s)
- Adrián Gómez-Baltazar
- División de Estudios de Posgrado, Facultad de Ciencias Médicas y Biológicas "Dr. Ignacio Chávez", Universidad Michoacana de San Nicolás de Hidalgo. Morelia, Michoacán, 58020, Mexico; Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo. Morelia, Michoacán, 58893, Mexico
| | - Gerardo Vázquez-Marrufo
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo. Morelia, Michoacán, 58893, Mexico
| | - Humberto Astiazaran-Garcia
- Departamento de Nutrición y Metabolismo, Centro de Investigación en Alimentación y Desarrollo, Carretera al Ejido "La Victoria" Km 0.6, 83304, Mexico
| | - Alejandra Ochoa-Zarzosa
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo. Morelia, Michoacán, 58893, Mexico
| | - Rafael Canett-Romero
- Departamento de Investigación y Posgrado en Alimentos, Universidad de Sonora, Hermosillo, Sonora, Mexico
| | - Alfonso García-Galaz
- Ciencias de los Alimentos, Centro de Investigación en Alimentación y Desarrollo AC, Carretera al Ejido La Victoria Km 0.6 CP 83304, Mexico
| | - Carlos Torres-Vega
- Laboratorio de Histología, Facultad de Ciencias Médicas y Biológicas "Dr. Ignacio Chávez", Universidad Michoacana de San Nicolás de Hidalgo. Morelia, Michoacán, 58020, Mexico
| | - Ma Soledad Vázquez-Garcidueñas
- División de Estudios de Posgrado, Facultad de Ciencias Médicas y Biológicas "Dr. Ignacio Chávez", Universidad Michoacana de San Nicolás de Hidalgo. Morelia, Michoacán, 58020, Mexico.
| |
Collapse
|
33
|
Boulanger EF, Sabag-Daigle A, Baniasad M, Kokkinias K, Schwieters A, Wrighton KC, Wysocki VH, Ahmer BMM. Sugar-Phosphate Toxicities Attenuate Salmonella Fitness in the Gut. J Bacteriol 2022; 204:e0034422. [PMID: 36383008 PMCID: PMC9765134 DOI: 10.1128/jb.00344-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 10/21/2022] [Indexed: 11/17/2022] Open
Abstract
Pathogens are becoming resistant to antimicrobials at an increasing rate, and novel therapeutic strategies are needed. Using Salmonella as a model, we have investigated the induction of sugar-phosphate toxicity as a potential therapeutic modality. The approach entails providing a nutrient while blocking the catabolism of that nutrient, resulting in the accumulation of a toxic intermediate. We hypothesize that this build-up will decrease the fitness of the organism during infection given nutrient availability. We tested this hypothesis using mutants lacking one of seven genes whose mutation is expected to cause the accumulation of a toxic metabolic intermediate. The araD, galE, rhaD, glpD, mtlD, manA, and galT mutants were then provided the appropriate sugars, either in vitro or during gastrointestinal infection of mice. All but the glpD mutant had nutrient-dependent growth defects in vitro, suggestive of sugar-phosphate toxicity. During gastrointestinal infection of mice, five mutants had decreased fitness. Providing the appropriate nutrient in the animal's drinking water was required to cause fitness defects with the rhaD and manA mutants and to enhance the fitness defect of the araD mutant. The galE and mtlD mutants were severely attenuated regardless of the nutrient being provided in the drinking water. Homologs of galE are widespread among bacteria and in humans, rendering the specific targeting of bacterial pathogens difficult. However, the araD, mtlD, and rhaD genes are not present in humans, appear to be rare in most phyla of bacteria, and are common in several genera of Enterobacteriaceae, making the encoded enzymes potential narrow-spectrum therapeutic targets. IMPORTANCE Bacterial pathogens are becoming increasingly resistant to antibiotics. There is an urgent need to identify novel drug targets and therapeutic strategies. In this work we have assembled and characterized a collection of mutations in our model pathogen, Salmonella enterica, that block a variety of sugar utilization pathways in such a way as to cause the accumulation of a toxic sugar-phosphate. Mutations in three genes, rhaD, araD, and mtlD, dramatically decrease the fitness of Salmonella in a mouse model of gastroenteritis, suggesting that RhaD, AraD, and MtlD may be good narrow-spectrum drug targets. The induction of sugar-phosphate toxicities may be a therapeutic strategy that is broadly relevant to other bacterial and fungal pathogens.
Collapse
Affiliation(s)
- Erin F. Boulanger
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Anice Sabag-Daigle
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Maryam Baniasad
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Katherine Kokkinias
- Department of Soil and Crop Science, Colorado State University, Ft. Collins, Colorado, USA
| | - Andrew Schwieters
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Kelly C. Wrighton
- Department of Soil and Crop Science, Colorado State University, Ft. Collins, Colorado, USA
| | - Vicki H. Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Brian M. M. Ahmer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
34
|
Rational consideration of Akkermansia muciniphila targeting intestinal health: advantages and challenges. NPJ Biofilms Microbiomes 2022; 8:81. [PMID: 36253412 PMCID: PMC9576740 DOI: 10.1038/s41522-022-00338-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 09/20/2022] [Indexed: 11/09/2022] Open
Abstract
As one of the promising next-generation probiotics (NGPs), Akkermansia muciniphila, a well-known mucin-degrading bacterium, has been proven to be closely related to the metabolic diseases of its human host. However, the role of A. muciniphila in the host’s intestinal health remains ambiguous. Here, we comprehensively summarize and discuss the characteristics, the distribution, and the colonization of A. muciniphila in the human gastrointestinal tract (GIT). We propose that the application of A. muciniphila as a biomarker for longevity, for diagnostics and prognostics of intestinal diseases, or for intestinal health should be cautiously considered. Precise dietary regulation can mediate the treatment of intestinal diseases by altering the abundance of A. muciniphila. Although the beneficial role of A. muciniphila and its component in intestinal inflammation has been discovered, in gnotobiotic mice with specific gut microbiota, certain genotype, and colorectal cancer, or in animal models infected with a specific pathogen, A. muciniphila may be related to the occurrence and development of intestinal diseases. Genomic analysis, emphasizing the strain-level phylogenetic differences of A. muciniphila, indicates that a clear description and discussion of each strain is critical before its practical application. Our review provides much needed insight for the precise application of A. muciniphila.
Collapse
|
35
|
Ge W, Zhou BG, Zhong YB, Liu SQ, Huang JQ, Yuan WY, Xie CY, Liu DY, Wang HY, Zuo ZY. Sishen Pill Ameliorates Dextran Sulfate Sodium (DSS)-Induced Colitis with Spleen-Kidney Yang Deficiency Syndromes: Role of Gut Microbiota, Fecal Metabolites, Inflammatory Dendritic Cells, and TLR4/NF- κB Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:6132289. [PMID: 36310616 PMCID: PMC9605852 DOI: 10.1155/2022/6132289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022]
Abstract
Sishen pill (SSP) is an old Chinese medicine used to treat colitis with spleen-kidney-yang deficiency (SKYD) syndromes. However, its exact mechanism of action has not yet been fully elucidated. The aim of this study was to evaluate the effects and potential mechanisms of SSP on colitis with SKYD syndromes in mice. Colitis with SKYD syndromes was induced by rhubarb, hydrocortisone, and dextran sulfate sodium (DSS), and treatment was provided with SSP. Flow cytometry was performed to examine the inflammatory dendritic cell (infDC) regulations of SSP. The changes in the gut microbiota (GM) and fecal metabolites post-SSP treatment were investigated using the combination of 16S rRNA sequencing and untargeted metabolomics. Additionally, we also examined whether SSPs could regulate the infDCs by modifying TLR4/NF-κB signaling pathways. Compared with the DSS group, the disease activity index, colonic weight, index of colonic weight, and colonic injury scores, as well as the levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and IL-12p70 decreased significantly in the DSS + SSP group, while free triiodothyronine (FT3), free tetraiodothyronine (FT4), testosterone (TESTO), body weight change, colonic length, and the levels of IL-10 increased. Also, SSP decreased the amounts of CD103+CD11c+iNOS+, CD103+CD11c+TNF-α +, CD11c+CD103+CD324+, CD103+CD11c+MHC-II+, and CD103+CD11c+CD115+. Interestingly, 16S rRNA sequencing and untargeted metabolomics showed that SSP treatment restored the dysbiosis of GM and improved the dysfunction in fecal metabolism in colitis mice with SKYD syndromes. Correlation analysis indicated that the modulatory effects of SSP on FT3, FT4, IL-10, colonic weight index, CD103+CD11c+TNF-α +, CD103+CD11c+MHC-II+, and 13 common differential metabolites were related to alterations in the abundance of Parvibacter, Aerococcus, norank_f_Lachnospiraceae, Lachnospiraceae_UCG-006, Akkermansia, and Rhodococcus in the GM. In addition, SSP markedly inhibited the activation of the TLR4, MyD88, TRAF6, TAB2, and NF-κBp65 proteins and activated IκB. These results indicate that SSP can effectively alleviate colitis mice with SKYD syndrome by regulating infDCs, GM, fecal metabolites, and TLR4/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Wei Ge
- Department of Proctology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, China
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Bu-Gao Zhou
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - You-Bao Zhong
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Su-Qing Liu
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jia-Qi Huang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Wang-Yuan Yuan
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Chang-Ying Xie
- Department of Proctology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Duan-Yong Liu
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Hai-Yan Wang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, China
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Zheng-Yun Zuo
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| |
Collapse
|
36
|
Wang S, Kang X, Alenius H, Wong SH, Karisola P, El-Nezami H. Oral exposure to Ag or TiO 2 nanoparticles perturbed gut transcriptome and microbiota in a mouse model of ulcerative colitis. Food Chem Toxicol 2022; 169:113368. [PMID: 36087619 DOI: 10.1016/j.fct.2022.113368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/05/2022] [Accepted: 08/10/2022] [Indexed: 11/28/2022]
Abstract
Silver (nAg) and titanium dioxide (nTiO2) nanoparticles improve texture, flavour or anti-microbial properties of various food products and packaging materials. Despite their increased oral exposure, their potential toxicities in the dysfunctional intestine are unclear. Here, the effects of ingested nAg or nTiO2 on inflamed colon were revealed in a mouse model of chemical-induced acute ulcerative colitis. Mice (eight/group) were exposed to nAg or nTiO2 by oral gavage for 10 consecutive days. We characterized disease phenotypes, histology, and alterations in colonic transcriptome (RNA sequencing) and gut microbiome (16S sequencing). Oral exposure to nAg caused only minor changes in phenotypic hallmarks of colitic mice but induced extensive responses in gene expression enriching processes of apoptotic cell death and RNA metabolism. Instead, ingested nTiO2 yielded shorter colon, aggravated epithelial hyperplasia and deeper infiltration of inflammatory cells. Both nanoparticles significantly changed the gut microbiota composition, resulting in loss of diversity and increase of potential pathobionts. They also increased colonic mucus and abundance of Akkermansia muciniphila. Overall, nAg and nTiO2 induce dissimilar immunotoxicological changes at the molecular and microbiome level in the context of colon inflammation. The results provide valuable information for evaluation of utilizing metallic nanoparticles in food products for the vulnerable population.
Collapse
Affiliation(s)
- Shuyuan Wang
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region of China.
| | - Xing Kang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China.
| | - Harri Alenius
- Human Microbiome Research Program, University of Helsinki, Haartmaninkatu 3, 00290, Helsinki, Finland; Institute of Environmental Medicine (IMM), Karolinska Institutet, Stockholm, 171 77, Sweden.
| | - Sunny Hei Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.
| | - Piia Karisola
- Human Microbiome Research Program, University of Helsinki, Haartmaninkatu 3, 00290, Helsinki, Finland.
| | - Hani El-Nezami
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region of China; Nutrition and Health, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
37
|
He Q, Shi Y, Xing H, Tang Q, Liu J, Li C, Zhang H, Zhang B, Zhang J, Chen X. Modulating effect of Xuanfei Baidu granule on host metabolism and gut microbiome in rats. Front Pharmacol 2022; 13:922642. [PMID: 36147334 PMCID: PMC9486314 DOI: 10.3389/fphar.2022.922642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Xuanfei Baidu granule (XFBD) is a recommended patented drug for the prevention and treatment of Corona Virus Disease 2019 (COVID-19), which is approved by the National Medical Products Administration. XFBD suppresses the over-activated immune response caused by inflammatory factor storms in COVID-19 infection. The intestine plays a crucial role in the immune system. The mass spectrometry based fecal metabolomics with 16S rDNA sequencing were combined to evaluate the effects of XFBD on host metabolism and gut microbiome. Short-chain fatty acids (SCFAs) contents in fecal matter were quantified by gas chromatography-mass spectrometry (GC-MS). Plasma samples were used to detect immune and inflammatory levels. The results were verified with a rat model of intestinal disorder. Results indicated that XFBD could increase the immune level of Immunoglobulin A (IgA), Immunoglobulin G (IgG) and Immunoglobulin M (IgM) (p < 0.05). The OPLS-DA analysis results showed that a total of 271 differential metabolites (178 up-regulated and 93 down-regulated) were identified based on the VIP ≥1, p < 0.05, FC ≥ 2 and FC ≤ 0.5. The metabolic pathways mainly involved D-Glutamine and D-glutamate metabolism, Arginine biosynthesis, Biotin metabolism, et al. XFBD modified the gut bacteria structure according to the principal component analysis (PCA), that is, 2 phyla, 3 classes, 5 orders, 11 families and 14 genera were significantly different based on taxonomic assignment. In addition, it could partially callback the relative abundance of intestinal microflora in bacterial disorder rats caused by antibiotics. It is suggested that the intervention mechanism of XFBD might be related to the regulation of intestinal flora composition. The evidence obtained in the study provides a useful reference for understanding the mechanism of XFBD.
Collapse
|
38
|
Herfindal AM, Rocha SDC, Papoutsis D, Bøhn SK, Carlsen H. The ROS-generating enzyme NADPH oxidase 1 modulates the colonic microbiota but offers minor protection against dextran sulfate sodium-induced low-grade colon inflammation in mice. Free Radic Biol Med 2022; 188:298-311. [PMID: 35752373 DOI: 10.1016/j.freeradbiomed.2022.06.234] [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: 05/11/2022] [Revised: 06/13/2022] [Accepted: 06/19/2022] [Indexed: 10/17/2022]
Abstract
The enzyme NADPH oxidase 1 (NOX1) is a major producer of superoxide which together with other reactive oxygen and nitrogen species (ROS/RNS) are implicated in maintaining a healthy epithelial barrier in the gut. While previous studies have indicated NOX1's involvement in microbial modulation in the small intestine, less is known about the effects of NOX1-dependent ROS/RNS formation in the colon. We investigated the role of NOX1 in the colon of NOX1 knockout (KO) and wild type (WT) mice, under mild and subclinical low-grade colon inflammation induced by 1% dextran sulfate sodium (DSS). Ex vivo imaging of ROS/RNS in the colon revealed that absence of NOX1 strongly decreased ROS/RNS production, particularly during DSS treatment. Furthermore, while absence of NOX1 did not affect disease activity, some markers of inflammation (mRNA: Tnfa, Il6, Ptgs2; protein: lipocalin 2) in the colonic mucosa tended to be higher in NOX1 KO than in WT mice following DSS treatment. Lack of NOX1 also extensively modulated the bacterial community in the colon (16S rRNA gene sequencing), where NOX1 KO mice were characterized mainly by lower α-diversity (richness and evenness), higher abundance of Firmicutes, Akkermansia, and Oscillibacter, and lower abundance of Bacteroidetes and Alistipes. Together, our data suggest that NOX1 is pivotal for colonic ROS/RNS production in mice both during steady-state (i.e., no DSS treatment) and during 1% DSS-induced low-grade inflammation and for modulation of the colonic microbiota, with potential beneficial consequences for intestinal health.
Collapse
Affiliation(s)
- Anne Mari Herfindal
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, P. O. Box 5003, N-1432, Ås, Norway.
| | - Sérgio Domingos Cardoso Rocha
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, P. O. Box 5003, N-1432, Ås, Norway; Faculty of Biosciences, Norwegian University of Life Sciences, P. O. Box 5003, N-1432, Ås, Norway.
| | - Dimitrios Papoutsis
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, P. O. Box 5003, N-1432, Ås, Norway.
| | - Siv Kjølsrud Bøhn
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, P. O. Box 5003, N-1432, Ås, Norway.
| | - Harald Carlsen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, P. O. Box 5003, N-1432, Ås, Norway.
| |
Collapse
|
39
|
Zhao X, Jiang L, Fang X, Guo Z, Wang X, Shi B, Meng Q. Host-microbiota interaction-mediated resistance to inflammatory bowel disease in pigs. MICROBIOME 2022; 10:115. [PMID: 35907917 PMCID: PMC9338544 DOI: 10.1186/s40168-022-01303-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 06/07/2022] [Indexed: 05/04/2023]
Abstract
BACKGROUND Disease resistance phenotypes are associated with immune regulatory functions and immune tolerance and have implications for both the livestock industry and human health. Microbiota plays an essential role in regulating immunity and autoimmunity in the host organism, but the influence of host-microbiota interactions on disease resistance phenotypes remains unclear. Here, multiomics analysis was performed to identify potential regulatory mechanisms of disease resistance at both the microbiome and host levels in two pig breeds. RESULTS Acute colitis models were established in Min pigs and Yorkshire pigs, and control and diseased individuals were compared. Compared with Yorkshire pigs under the same nutritional and management conditions, Min pigs exhibited strong disease resistance, as indicated by a low disease activity index (DAI) and a low histological activity index (HAI). Microbiota sequencing analysis showed that potentially harmful microbes Desulfovibrio, Bacteroides and Streptococcus were enriched in diseased individuals of the two breeds. Notably, potentially beneficial microbes, such as Lactobacillus, Clostridia and Eubacterium, and several genera belonging to Ruminococcaceae and Christensenellaceae were enriched in diseased Min pigs and were found to be positively associated with the microbial metabolites related to intestinal barrier function. Specifically, the concentrations of indole derivatives and short-chain fatty acids were increased in diseased Min pigs, suggesting beneficial action in protecting intestinal barrier. In addition, lower concentrations of bile acid metabolites and short-chain fatty acids were observed in diseased Yorkshire pigs, which were associated with increased potentially harmful microbes, such as Bilophila and Alistipes. Concerning enrichment of the immune response, the increase in CD4+ T cells in the lamina propria improved supervision of the host immunity response in diseased Min pigs, contributing to the maintenance of Th2-type immune superiority and immune tolerance patterns and control of excessive inflammation with the help of potentially beneficial microbes. In diseased Yorkshire pigs, more terms belonging to biological processes of immunity were enriched, including Toll-like receptors signalling, NF-κB signalling and Th1 and Th17-type immune responses, along with the increases of potentially harmful microbes and damaged intestinal barrier. CONCLUSIONS Cumulatively, the results for the two pig breeds highlight that host-microbiota crosstalk promotes a disease resistance phenotype in three ways: by maintaining partial PRR nonactivation, maintaining Th2-type immune superiority and immunological tolerance patterns and recovering gut barrier function to protect against colonic diseases. Video abstract.
Collapse
Affiliation(s)
- Xuan Zhao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Lin Jiang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xiuyu Fang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Zhiqiang Guo
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xiaoxu Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Baoming Shi
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| | - Qingwei Meng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
40
|
Huang X, Gao Y, Chen W, Hu Q, He Z, Wang X, Li D, Lin R. Dietary variety relates to gut microbiota diversity and abundance in humans. Eur J Nutr 2022; 61:3915-3928. [PMID: 35764724 DOI: 10.1007/s00394-022-02929-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/31/2022] [Indexed: 02/08/2023]
Abstract
PURPOSE We aim to investigate the relationship between gut microbiota and dietary variety in a Chinese population using Dietary Variety Score (DVS), an index of dietary variety, as little has studied the relationship of dietary variety and gut microbiota in a general population. METHODS In this cross-sectional study, recruited participants were conducted with face-to-face interview to collect information on 24-h food intake and dietary consumption using a valid food frequency questionnaire. Subjects (n = 128) were divided as high and low DVS groups by the median of DVS after rigorously matching for confounding factors. The gut microbiota was assessed by 16S rRNA sequencing and the correlations between key phylotypes and DVS, Index of Nutritional Quality (INQ) and clinical indices were examined using generalized linear model in negative binomial regression. RESULTS Higher score of DVS, INQVB6, INQVE and INQZn exhibited higher α-diversity. DVS was correlated with INQ and six genera. Among the DVS-correlated genera, Turicibacter, Alistipes and Barnesiella were positively correlated with INQVE, INQZn and INQCu, individually or in combination, while Cetobacterium was negatively correlated with INQCu, INQZn and INQVE. The abundance of Coprococcus and Barnesiella increased with the elevated cumulative scores of INQVE, INQVB6 and INQZn. The combination of Alistipes, Roseburia and Barnesiella could moderately predict dietary variety status. CONCLUSION Higher DVS was correlated with higher microbial diversity and more abundance of some potentially beneficial bacteria but with less some potentially pathogenic bacteria. A high variety dietary, therefore, should be recommended in our daily life.
Collapse
Affiliation(s)
- Xueran Huang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, China.,Department of Nutrition and Food Hygiene, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Yongfen Gao
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, China.,Department of Nutrition and Food Hygiene, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Wanrong Chen
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, China.,Department of Nutrition and Food Hygiene, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Qiantu Hu
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, China
| | - Zouyan He
- Department of Nutrition and Food Hygiene, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Xi Wang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, China.,Department of Nutrition and Food Hygiene, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Dan Li
- The Third Affiliated Hospital of Guangxi Medical University, Nanning Second Peoples Hospital, Nanning, 530031, China.
| | - Rui Lin
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, China. .,Department of Nutrition and Food Hygiene, School of Public Health, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
41
|
Liu D, Zhao R, Wu Y, Wang Y, Yang R, Ke X. Variation in the Efficacy of Anti-Ulcerative Colitis Treatments Reveals the Conflict Between Precipitating Compatibility of Traditional Chinese Medicine and Modern Technology: A Case of Scutellaria-Coptis. Front Pharmacol 2022; 13:819851. [PMID: 35517805 PMCID: PMC9065555 DOI: 10.3389/fphar.2022.819851] [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: 11/22/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Scutellariae and Coptidis compose a classical drug pair applied in clinical practice to dispel heat, dryness, and dampness, and they are also precipitation compatible drug pairs. With modern technology, Scutellaria-Coptis is mostly prepared by decocting its components separately, while in the traditional method, it is predominantly prepared as a combined decoction. The present study investigated the effects and mechanisms of separate and combined application of Scutellaria-Coptis decoction on ulcerative colitis (UC) in mice induced by the administration of dextran sulfate sodium (DSS). Changes in body weight, colon length, and Disease Activity Index scores were also evaluated. Hematoxylin and eosin staining and other methods were used to evaluate the overall condition of animals in each group. Intestinal microflora was analyzed using 16S rRNA sequencing, while colon inflammation and antioxidant capacity were evaluated based on the levels of interleukin-6 (IL-6), IL-10, IL-1β, tumor necrosis factor-α, superoxide dismutase, malondialdehyde, and reduced glutathione. The results revealed that Scutellaria-Coptis significantly relieved colon inflammation in mice, and the combined decoction of Scutellaria-Coptis exerted a significant effect on UC. Notably, the protective effect of Scutellaria-Coptis against colon inflammation was weakened when the antibiotic mixture was partially consumed by the gut microbiota. The results of 16S rRNA sequencing showed that the group treated with combined decoction of Scutellaria-Coptis exhibited a higher intestinal microbial diversity and intestinal flora composition than the separated decoction group. Treatment of mice with UC by administering Scutellaria-Coptis decoction through intestinal flora removal (ABX) and fecal microbial transplantation (FMT) was closely associated with intestinal flora composition. In conclusion, Scutellaria-Coptis can relieve UC with an excellent effect especially when taken as a combined decoction, alleviating colon inflammation incurred by intestinal microbes to a certain extent.
Collapse
Affiliation(s)
- Dan Liu
- Chongqing Key Laboratory of Chinese Medicine New Drug Screening, Southwest University, Chongqing, China
| | - Ran Zhao
- Chongqing Key Laboratory of Chinese Medicine New Drug Screening, Southwest University, Chongqing, China
| | - Yajing Wu
- Chongqing Key Laboratory of Chinese Medicine New Drug Screening, Southwest University, Chongqing, China
| | - Yunhong Wang
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Rongping Yang
- Chongqing Key Laboratory of Chinese Medicine New Drug Screening, Southwest University, Chongqing, China
| | - Xiumei Ke
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China.,School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
| |
Collapse
|
42
|
Nadimpalli ML, Lanza VF, Montealegre MC, Sultana S, Fuhrmeister ER, Worby CJ, Teichmann L, Caduff L, Swarthout JM, Crider YS, Earl AM, Brown J, Luby SP, Islam MA, Julian TR, Pickering AJ. Drinking water chlorination has minor effects on the intestinal flora and resistomes of Bangladeshi children. Nat Microbiol 2022; 7:620-629. [PMID: 35422497 PMCID: PMC9249080 DOI: 10.1038/s41564-022-01101-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 02/24/2022] [Indexed: 02/07/2023]
Abstract
Healthy development of the gut microbiome provides long-term health benefits. Children raised in countries with high infectious disease burdens are frequently exposed to diarrhoeal pathogens and antibiotics, which perturb gut microbiome assembly. A recent cluster-randomized trial leveraging >4,000 child observations in Dhaka, Bangladesh, found that automated water chlorination of shared taps effectively reduced child diarrhoea and antibiotic use. In this substudy, we leveraged stool samples collected from 130 children 1 year after chlorine doser installation to examine differences between treatment and control children's gut microbiota. Water chlorination was associated with increased abundance of several bacterial genera previously linked to improved gut health; however, we observed no effects on the overall richness or diversity of taxa. Several clinically relevant antibiotic resistance genes were relatively more abundant in the gut microbiome of treatment children, possibly due to increases in Enterobacteriaceae. While further studies on the long-term health impacts of drinking chlorinated water would be valuable, we conclude that access to chlorinated water did not substantially impact child gut microbiome development in this setting, supporting the use of chlorination to increase global access to safe drinking water.
Collapse
Affiliation(s)
- Maya L. Nadimpalli
- Department of Civil and Environmental Engineering, Tufts University, Medford, MA, USA,Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance (Levy CIMAR), Tufts University, Boston, MA, USA
| | - Val F. Lanza
- Bioinformatics Unit, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain,Network Research Center for Infectious Diseases (CIBERINFEC), Spain
| | | | - Sonia Sultana
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Erica R. Fuhrmeister
- Department of Civil and Environmental Engineering, Tufts University, Medford, MA, USA
| | - Colin J. Worby
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA
| | - Lisa Teichmann
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Lea Caduff
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland
| | - Jenna M. Swarthout
- Department of Civil and Environmental Engineering, Tufts University, Medford, MA, USA
| | - Yoshika S. Crider
- Energy and Resources Group, University of California Berkeley, Berkeley, CA, USA,King Center on Global Development, Stanford University, Stanford, CA, USA
| | - Ashlee M. Earl
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA
| | - Joe Brown
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Stephen P. Luby
- Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| | - Mohammad Aminul Islam
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland,Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Timothy R. Julian
- Network Research Center for Infectious Diseases (CIBERINFEC), Spain,Swiss Tropical and Public Health Institute, Basel, Switzerland,University of Basel, Basel, Switzerland
| | - Amy J. Pickering
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance (Levy CIMAR), Tufts University, Boston, MA, USA,Department of Civil and Environmental Engineering, University of California, Berkeley, CA, USA,Blum Center for Developing Economies, University of California, Berkeley, CA, USA,Correspondence and requests for materials should be addressed to Dr. Amy Pickering () and Dr. Tim Julian ()
| |
Collapse
|
43
|
Wong EOY, Brownlie EJE, Ng KM, Kathirgamanathan S, Yu FB, Merrill BD, Huang KC, Martin A, Tropini C, Navarre WW. The CIAMIB: a Large and Metabolically Diverse Collection of Inflammation-Associated Bacteria from the Murine Gut. mBio 2022; 13:e0294921. [PMID: 35266814 PMCID: PMC9040815 DOI: 10.1128/mbio.02949-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/09/2022] [Indexed: 02/07/2023] Open
Abstract
Gut inflammation directly impacts the growth and stability of commensal gut microbes and can lead to long-lasting changes in microbiota composition that can prolong or exacerbate disease states. While mouse models are used extensively to investigate the interplay between microbes and the inflamed state, the paucity of cultured mouse gut microbes has hindered efforts to determine causal relationships. To address this issue, we are assembling the Collection of Inflammation-Associated Mouse Intestinal Bacteria (CIAMIB). The initial release of this collection comprises 41 isolates of 39 unique bacterial species, covering 4 phyla and containing 10 previously uncultivated isolates, including 1 novel family and 7 novel genera. The collection significantly expands the number of available Muribaculaceae, Lachnospiraceae, and Coriobacteriaceae isolates and includes microbes from genera associated with inflammation, such as Prevotella and Klebsiella. We characterized the growth of CIAMIB isolates across a diverse range of nutritional conditions and predicted their metabolic potential and anaerobic fermentation capacity based on the genomes of these isolates. We also provide the first metabolic analysis of species within the genus Adlercreutzia, revealing these representatives to be nitrate-reducing and severely restricted in their ability to grow on carbohydrates. CIAMIB isolates are fully sequenced and available to the scientific community as a powerful tool to study host-microbiota interactions. IMPORTANCE Attempts to explore the role of the microbiota in animal physiology have resulted in large-scale efforts to cultivate the thousands of microbes that are associated with humans. In contrast, relatively few lab mouse-associated bacteria have been isolated, despite the fact that the overwhelming number of studies on the microbiota use laboratory mice that are colonized with microbes that are quite distinct from those in humans. Here, we report the results of a large-scale isolation of bacteria from the intestines of laboratory mice either prone to or suffering from gut inflammation. This collection comprises dozens of novel isolates, many of which represent the only cultured representatives of their genus or species. We report their basic growth characteristics and genomes and are making them widely available to the greater research community.
Collapse
Affiliation(s)
- Erin Oi-Yan Wong
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Katharine Michelle Ng
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | | | | | - Bryan D. Merrill
- Department of Microbiology and Immunology, Stanford University of School of Medicine, Stanford, California, USA
| | - Kerwyn Casey Huang
- Chan Zuckerberg Biohub, San Francisco, California, USA
- Department of Microbiology and Immunology, Stanford University of School of Medicine, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Alberto Martin
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Carolina Tropini
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, Canada
| | | |
Collapse
|
44
|
Bai X, Shi Y, Tang L, Chen L, Fan H, Wang H, Wang J, Jia X, Chen S, Lai S. Heat Stress Affects Faecal Microbial and Metabolic Alterations of Rabbits. Front Microbiol 2022; 12:817615. [PMID: 35295680 PMCID: PMC8919919 DOI: 10.3389/fmicb.2021.817615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 12/31/2021] [Indexed: 12/19/2022] Open
Abstract
Heat stress can impair the rabbit immune system, induce oxidative stress, and cause many complications. These diseases are characterized by metabolic disorders, but the underlying mechanism is unknown. As a result, the current research determines the effects of HS on intestinal microorganisms in rabbits and the metabolic pathway disorders caused by HS. Twelve rabbits were randomly assigned to one of two groups: CON (22–24°C) and HS (30°C–32°C). Both the groups were treated for 15 days. Blood and fecal samples were collected on day 15. Serum immune oxidation indices were determined using a commercial ELISA kit, and the microbiome of rabbit feces was studied using 16S rRNA gene sequencing. Non-targeted metabolomics was analyzed using ultra-high-performance liquid chromatography-mass spectrometry (UHPC MS/MS). The findings revealed that HS significantly increased IgG and T-AOC levels in serum, whereas it decreased TNF-α and IL-10. NMDS analysis revealed a substantial difference in bacterial community composition between HS and CON groups. At the phylum level, the abundance of Firmicutes, Protobacteria, and Verrucomicrobiota was significantly higher in the HS group, whereas the abundance of Bacteriodota was reduced in the CON group. V9D2013 group, Haloplasma, Comamonas, Clostridium sensu stricto 1, Ruminiclostridium, Syntrophus Lutispora, at the genus level Syntrophorhabdus, Paeniclostridium, Clostridium sensu stricto 6, Candidatus Caldatribacterium, Spirochaeta Synergistaceae, Syner-01, [Eubacterium] xylanophilum group, Cellulosilyticum, ADurb.Bin120, and Devosia were significantly upregulated in the HS group. The metabolism of the HS group was considerably upregulated compared with the metabolism of the CON group, according to principal component analysis (PCA) and least-squares discriminant analysis (PLS-DA). HS increased the concentrations of 4-pyridoxic acid, kynurenine, 20-OH-leukotriene B4, and dopamine and decreased the concentration of pyridoxal. In the rabbit gut, these compounds primarily impact the metabolic pathways of vitamin B6, tryptophan, neutrophil activation, and prolactin. 4-Pyridoxic acid, pyridoxal, kynurenine, 20-OH-leukotriene B4, and dopamine are essential inflammatory response markers and oxidative stress.
Collapse
Affiliation(s)
- Xue Bai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yu Shi
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Lipeng Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Li Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Huimei Fan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Haoding Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jie Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xianbo Jia
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Shiyi Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Songjia Lai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
45
|
Yang K, Deng X, Jian S, Zhang M, Wen C, Xin Z, Zhang L, Tong A, Ye S, Liao P, Xiao Z, He S, Zhang F, Deng J, Zhang L, Deng B. Gallic Acid Alleviates Gut Dysfunction and Boosts Immune and Antioxidant Activities in Puppies Under Environmental Stress Based on Microbiome-Metabolomics Analysis. Front Immunol 2022; 12:813890. [PMID: 35095912 PMCID: PMC8795593 DOI: 10.3389/fimmu.2021.813890] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Early-life exposure to environmental stress disrupts the gut barrier and leads to inflammatory responses and changes in gut microbiota composition. Gallic acid (GA), a natural plant polyphenol, has received significant interest for its antioxidant, anti-inflammatory, and antimicrobial properties that support the maintenance of intestinal health. To assess whether dietary supplementation of GA alleviates environmental stress, a total of 19 puppies were randomly allocated to the following three dietary treatments for 2 weeks: 1) basal diet (control (CON)); 2) basal diet + transportation (TS); and 3) basal diet with the addition of 500 mg/kg of GA + transportation (TS+GA). After a 1-week supplementation period, puppies in the TS and TS+GA groups were transported from a stressful environment to another livable location, and puppies in the CON group were then left in the stressful environment. Results indicated that GA markedly reduced the diarrhea rate in puppies throughout the trial period and caused a moderate decline of serum cortisol and HSP-70 levels after transportation. Also, GA alleviated the oxidative stress and inflammatory response caused by multiple environmental stressors. Meanwhile, puppies fed GA had a higher abundance of fecal Firmicutes and Lactobacillus and lower Proteobacteria, Escherichia–Shigella, and Clostridium_sensu_stricto_1 after transportation. As a result, the TS+GA group had the highest total short-chain fatty acids and acetic acid. Also, the fecal and serum metabolomics analyses revealed that GA markedly reversed the abnormalities of amino acid metabolism, lipid metabolism, carbohydrate metabolism, and nucleotide metabolism caused by stresses. Finally, Spearman’s correlation analysis was carried out to explore the comprehensive microbiota and metabolite relationships. Overall, dietary supplementation of GA alleviates oxidative stress and inflammatory response in stressed puppies by causing beneficial shifts on gut microbiota and metabolites that may support gut and host health.
Collapse
Affiliation(s)
- Kang Yang
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaolin Deng
- Department of Urology, Ganzhou People's Hospital, Ganzhou, China
| | - Shiyan Jian
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Meiyu Zhang
- College of Animal Science and Technology, Guangdong Polytechnic of Science and Trade, Guangzhou, China
| | - Chaoyu Wen
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhongquan Xin
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Limeng Zhang
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Aorigeile Tong
- Research Center of Pet Nutrition, Guangzhou Qingke Biotechnology Co., Ltd., Guangzhou, China
| | - Shibin Ye
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Pinfeng Liao
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zaili Xiao
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shansong He
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Fan Zhang
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jinping Deng
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Lingna Zhang
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Baichuan Deng
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
46
|
Baniasad M, Kim Y, Shaffer M, Sabag-Daigle A, Leleiwi I, Daly RA, Ahmer BMM, Wrighton KC, Wysocki VH. Optimization of proteomics sample preparation for identification of host and bacterial proteins in mouse feces. Anal Bioanal Chem 2022; 414:2317-2331. [PMID: 35106611 PMCID: PMC9393048 DOI: 10.1007/s00216-022-03885-z] [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: 09/22/2021] [Revised: 12/03/2021] [Accepted: 01/07/2022] [Indexed: 11/01/2022]
Abstract
Bottom-up proteomics is a powerful method for the functional characterization of mouse gut microbiota. To date, most of the bottom-up proteomics studies of the mouse gut rely on limited amounts of fecal samples. With mass-limited samples, the performance of such analyses is highly dependent on the protein extraction protocols and contaminant removal strategies. Here, protein extraction protocols (using different lysis buffers) and contaminant removal strategies (using different types of filters and beads) were systematically evaluated to maximize quantitative reproducibility and the number of identified proteins. Overall, our results recommend a protein extraction method using a combination of sodium dodecyl sulfate (SDS) and urea in Tris-HCl to yield the greatest number of protein identifications. These conditions led to an increase in the number of proteins identified from gram-positive bacteria, such as Firmicutes and Actinobacteria, which is a challenging task. Our analysis further confirmed these conditions led to the extraction of non-abundant bacterial phyla such as Proteobacteria. In addition, we found that, when coupled to our optimized extraction method, suspension trap (S-Trap) outperforms other contaminant removal methods by providing the most reproducible method while producing the greatest number of protein identifications. Overall, our optimized sample preparation workflow is straightforward and fast, and requires minimal sample handling. Furthermore, our approach does not require high amounts of fecal samples, a vital consideration in proteomics studies where mice produce smaller amounts of feces due to a particular physiological condition. Our final method provides efficient digestion of mouse fecal material, is reproducible, and leads to high proteomic coverage for both host and microbiome proteins.
Collapse
Affiliation(s)
- Maryam Baniasad
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Yongseok Kim
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Michael Shaffer
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO, USA
| | - Anice Sabag-Daigle
- Department of Microbiology, The Ohio State University, Columbus, OH, USA
| | - Ikaia Leleiwi
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO, USA
| | - Rebecca A Daly
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO, USA
| | - Brian M M Ahmer
- Department of Microbiology, The Ohio State University, Columbus, OH, USA
| | - Kelly C Wrighton
- Department of Soil and Crop Sciences, The Colorado State University, Fort Collins, CO, USA
| | - Vicki H Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
47
|
Could Dietary Supplementation with Different Sources of N-3 Polyunsaturated Fatty Acids Modify the Rabbit Gut Microbiota? Antibiotics (Basel) 2022; 11:antibiotics11020227. [PMID: 35203829 PMCID: PMC8868100 DOI: 10.3390/antibiotics11020227] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 11/22/2022] Open
Abstract
The present study evaluated the effects of feed supplemented with two dietary sources of n-3 polyunsaturated fatty acids (PUFAs; fish oil and extruded flaxseed) on the gut microbiota, caecal fermentations, gastrointestinal histology, and histochemistry in rabbits. Fifteen male New Zealand White rabbits were divided into three groups (n = 5/group) and fed with different diets from weaning (35 days of age) until slaughtering (90 days of age): C group, fed with a commercial diet; F group, supplemented with 10% of extruded flaxseed; and O group, supplemented with 3.5% of fish oil. At slaughter, the content of the stomach, duodenum, jejunum, ileum, caecum, and colon was collected and analyzed by Next Generation 16S rRNA gene sequencing. Tissue samples of the same tracts were evaluated with histological and histochemical analysis. Ammonia and lactic acid in the caecum were also quantified. Twenty-nine operational taxonomic units (OTUs) were significantly different between groups. Groups receiving n-3 PUFAs supplementation showed an increase in Bacteroidetes and Lachnospiraceae in several gastrointestinal tracts, while Bacilli abundance, as well as Firmicutes/Bacteroidetes ratio, were reduced compared to the control group (for all p < 0.05). Caecal ammonia was lower in the F than C group (p < 0.032), whereas no difference was found for lactic acid. Finally, histological evaluations revealed a mild hemorrhagic infiltration and vessels ectasia in the stomach mucosa of both F and O groups, but no effect of nutritional treatment was evidenced by the histochemical analyses. In conclusion, n-3 PUFAs supplementation could modify the rabbit gut microbiota and fermentation. The increase in beneficial bacterial populations may, at least partially, explain the positive effects of n-3 PUFAs diet supplementation on human and animals’ health, although the appropriate dosage should be established.
Collapse
|
48
|
Yuan X, Xue H, Xu X, Jiao X, Pan Z, Zhang Y. Closely related Salmonella Derby strains triggered distinct gut microbiota alteration. Gut Pathog 2022; 14:6. [PMID: 35078518 PMCID: PMC8787955 DOI: 10.1186/s13099-022-00480-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 01/13/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Salmonella Derby is one of the most predominant Salmonella serotypes that seriously threatens food safety. This bacterium can be further differentiated to sub-populations with different population sizes; however, whether and how the S. Derby–gut microbiota interactions affect epidemic patterns of S. Derby sub-populations remain largely unknown.
Results
We selected two representative strains, 14T and 14C, which represent rarely distributed and prevalent sub-populations of the S. Derby ST40 group, respectively, to address this question using a mouse model. Effects of oral administration of both strains was monitored for 14 days. Alpha diversity of gut microbiota at early stages of infection (4 h post infection) was higher in 14C-treated mice and lower in 14T-treated mice compared with controls. Strain 14T triggered stronger inflammation responses but with lower pathogen titer in spleen compared with strain 14C at 14 days post infection. Certain known probiotic bacteria that can hinder colonization of Salmonella, such as Bifidobacteriaceae and Akkermansiaceae, exhibited increased relative abundance in 14T-treated mice compared with 14C-treated mice. Our results also demonstrated that Ligilactobacillus strains isolated from gut microbiota showed stronger antagonistic activity against strain 14T compared with strain 14C.
Conclusions
We identified how S. Derby infection affected gut microbiota composition, and found that the 14T strain, which represented a rarely distributed S. Derby sub-population, triggered stronger host inflammation responses and gut microbiota disturbance compared with the 14C strain, which represented a prevalent S. Derby sub-population. This study provides novel insights on the impacts of gut microbiota on the epidemic patterns of Salmonella populations.
Collapse
|
49
|
Shi Y, Zou Y, Xiong Y, Zhang S, Song M, An X, Liu C, Zhang W, Chen S. Host Gasdermin D restrains systemic endotoxemia by capturing Proteobacteria in the colon of high-fat diet-feeding mice. Gut Microbes 2021; 13:1946369. [PMID: 34275417 PMCID: PMC8288038 DOI: 10.1080/19490976.2021.1946369] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gasdermin D (GSDMD) functions as a key pyroptotic executor through its secreted N-terminal domain (GSDMD-N). However, the functional relevance and mechanistic basis of the precise roles of host colonic GSDMD in high-fat diet (HFD)-induced gut dysbiosis and systemic endotoxemia remain elusive. In this study, we demonstrate that HFD feeding triggers GSDMD-N secretion of both T-lymphocytes and enterocytes in mouse colons. GSDMD deficiency aggravates HFD-induced systemic endotoxemia, gut barrier impairment, and colonic inflammation. More importantly, active GSDMD-N kills the Proteobacteria phylum via directly interacting with Cardiolipin. Mechanistically, we identify that the Glu236 (a known residue for GSDMD protein cleavage) is a bona fide important site for the bacterial recognition of GSDMD. Collectively, our findings explain the mechanism by which colonic GSDMD-N maintains low levels of HFD-induced metabolic endotoxemia. A GSDMD-N mimetic containing an exposed Glu236 site could be an attractive strategy for the treatment of HFD-induced metabolic endotoxemia.
Collapse
Affiliation(s)
- Yujie Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yixin Zou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yonghong Xiong
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shiyao Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China,Wenxiang Zhang State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Avenue, Nanjing211198, China
| | - Mingming Song
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaofei An
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenxiang Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China,Wenxiang Zhang State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Avenue, Nanjing211198, China
| | - Siyu Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China,CONTACT Siyu Chen
| |
Collapse
|
50
|
McLaughlin MC, McDevitt B, Miller H, Amundson KK, Wilkins MJ, Warner NR, Blotevogel J, Borch T. Constructed wetlands for polishing oil and gas produced water releases. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2021; 23:1961-1976. [PMID: 34723304 DOI: 10.1039/d1em00311a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Produced water (PW) is the largest waste stream associated with oil and gas (O&G) operations and contains petroleum hydrocarbons, heavy metals, salts, naturally occurring radioactive materials and any remaining chemical additives. In some areas in Wyoming, constructed wetlands (CWs) are used to polish PW downstream of National Pollutant Discharge Elimination System (NPDES) PW release points. In recent years, there has been increased interest in finding lower cost options, such as CWs, for PW treatment. The goal of this study was to understand the efficacy of removal and environmental fate of O&G organic chemical additives in CW systems used to treat PW released for agricultural beneficial reuse. To achieve this goal, we analyzed water and sediment samples for organic O&G chemical additives and conducted 16S rRNA gene sequencing for microbial community characterization on three such systems in Wyoming, USA. Three surfactants (polyethylene glycols, polypropylene glycols, and nonylphenol ethoxylates) and one biocide (alkyldimethylammonium chloride) were detected in all three PW discharges and >94% removal of all species from PW was achieved after treatment in two CWs in series. These O&G extraction additives were detected in all sediment samples collected downstream of PW discharges. Chemical and microbial analyses indicated that sorption and biodegradation were the main attenuation mechanisms for these species. Additionally, all three discharges showed a trend of increasingly diverse, but similar, microbial communities with greater distance from NPDES PW discharge points. Results of this study can be used to inform design and management of constructed wetlands for produced water treatment.
Collapse
Affiliation(s)
- Molly C McLaughlin
- Department of Civil and Environmental Engineering, Colorado State University, 1320 Campus Delivery, Fort Collins, CO, 80523, USA.
| | - Bonnie McDevitt
- Department of Civil and Environmental Engineering, The Pennsylvania State University, 212 Sackett Building, University Park, PA 16801, USA
| | - Hannah Miller
- Department of Soil and Crop Sciences, Colorado State University, 1170 Campus Delivery, Fort Collins, Colorado 80523, USA
| | - Kaela K Amundson
- Department of Soil and Crop Sciences, Colorado State University, 1170 Campus Delivery, Fort Collins, Colorado 80523, USA
| | - Michael J Wilkins
- Department of Soil and Crop Sciences, Colorado State University, 1170 Campus Delivery, Fort Collins, Colorado 80523, USA
| | - Nathaniel R Warner
- Department of Civil and Environmental Engineering, The Pennsylvania State University, 212 Sackett Building, University Park, PA 16801, USA
| | - Jens Blotevogel
- Department of Civil and Environmental Engineering, Colorado State University, 1320 Campus Delivery, Fort Collins, CO, 80523, USA.
| | - Thomas Borch
- Department of Civil and Environmental Engineering, Colorado State University, 1320 Campus Delivery, Fort Collins, CO, 80523, USA.
- Department of Soil and Crop Sciences, Colorado State University, 1170 Campus Delivery, Fort Collins, Colorado 80523, USA
- Department of Chemistry, Colorado State University, 1872 Campus Delivery, Fort Collins, Colorado, 80523, USA
| |
Collapse
|