1
|
Zhao X, Qiu Y, Liang L, Fu X. Interkingdom signaling between gastrointestinal hormones and the gut microbiome. Gut Microbes 2025; 17:2456592. [PMID: 39851261 PMCID: PMC11776477 DOI: 10.1080/19490976.2025.2456592] [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: 05/12/2024] [Revised: 08/12/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
The interplay between the gut microbiota and gastrointestinal hormones plays a pivotal role in the health of the host and the development of diseases. As a vital component of the intestinal microecosystem, the gut microbiota influences the synthesis and release of many gastrointestinal hormones through mechanisms such as modulating the intestinal environment, producing metabolites, impacting mucosal barriers, generating immune and inflammatory responses, and releasing neurotransmitters. Conversely, gastrointestinal hormones exert feedback regulation on the gut microbiota by modulating the intestinal environment, nutrient absorption and utilization, and the bacterial biological behavior and composition. The distributions of the gut microbiota and gastrointestinal hormones are anatomically intertwined, and close interactions between the gut microbiota and gastrointestinal hormones are crucial for maintaining gastrointestinal homeostasis. Interventions leveraging the interplay between the gut microbiota and gastrointestinal hormones have been employed in the clinical management of metabolic diseases and inflammatory bowel diseases, such as bariatric surgery and fecal microbiota transplantation, offering promising targets for the treatment of dysbiosis-related diseases.
Collapse
Affiliation(s)
- Xinyu Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Ye Qiu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Lanfan Liang
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiangsheng Fu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Wang J, Zhang Q, Wang H, Liu C, Jiang L, Liu W, Wu Y, Wang Y, Vivian, Yan H, Lin J, Sun X. A Sr@Ag-based spatiotemporal and step-release scaffold against chronic osteomyelitis, fabricated by coaxial 3D-printing. Biomaterials 2025; 314:122899. [PMID: 39437580 DOI: 10.1016/j.biomaterials.2024.122899] [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: 04/09/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
Chronic osteomyelitis (OM) represents a severe and persistent infectious bone disease. Effective treatment requires controlled anti-inflammatory releases and bone regeneration across disease phases. A Sr@Ag-based scaffold was successfully printed, featuring micron-scale coaxial fibers containing Ag-doped hydroxyapatite (HA) in the outer layer of PLLA and Sr-doped HA in the inner layer of PLLA, facilitating the spatiotemporal and sequential release of Ag and Sr ions during OM treatment. Most antibacterial agent (Ag) was released during the first 20 days, followed by a slow-release plateau over the next 40 days in phosphate-buffered saline solution (PBS). Meanwhile, the pro-angiogenic agent (Sr) was released in minimal amounts during the initial 20 days, followed by a rapid and considerable release in the following 40 days. The coaxial design effectively inhibited the growth of Staphylococcus aureus and Escherichia coli while preserving the viability of bone cells. The ion-based scaffold exhibited broad-spectrum antibacterial effects and enhanced bone-regenerating gene expression in a complex air-bacteria environment. The Sr@Ag-based coaxial scaffold demonstrated effective antibacterial activity during the early stage and exhibited excellent non-toxic bone regeneration results during the middle and late stages in vivo. This work offered a promising treatment strategy through sequential anti-inflammatory and pro-osteogenic effects for infectious bone-defect diseases.
Collapse
Affiliation(s)
- Jingyun Wang
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Qin Zhang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, Jiangsu, 215000, People's Republic of China; Department of Orthopaedics, First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, 215000, People's Republic of China
| | - Hetong Wang
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Chunlin Liu
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; School of Mechanical Engineering, Chengdu University, Chengdu, Sichuan, 610000, People's Republic of China.
| | - Le Jiang
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Wanting Liu
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Yixian Wu
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Yifan Wang
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Vivian
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Hao Yan
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, People's Republic of China
| | - Jun Lin
- Department of Orthopaedics, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, Jiangsu, 215000, People's Republic of China; Department of Orthopaedics, First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, 215000, People's Republic of China.
| | - Xiaodan Sun
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China.
| |
Collapse
|
3
|
Ruiz A, Gisbert E, Andree KB. Impact of the diet in the gut microbiota after an inter-species microbial transplantation in fish. Sci Rep 2024; 14:4007. [PMID: 38369563 PMCID: PMC10874947 DOI: 10.1038/s41598-024-54519-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/13/2024] [Indexed: 02/20/2024] Open
Abstract
Inter-species microbial transplantations offer the possibility of transferring species-specific microbes and their associated functionality. As a conceptual approach, an intestinal microbiota transplant (IMT) between two marine carnivorous fish species that thrive in different environmental conditions was conducted: from donor Atlantic salmon (Salmo salar) to recipient gilthead seabream (Sparus aurata), after obliterating its basal microbiota with an antibiotic treatment. To confirm that the gut microbiota was able to recover after antibiotics without the influence of the diet, a group of gilthead seabream not submitted to the IMT was kept fasted as an internal control. To assess the effect of the diet after the IMT, two groups of gilthead seabream were respectively fed with their typical diet and with Atlantic salmon diet. At 36 days post-IMT, the gut of the individuals fed with their typical diet was dominated by the feed-associated bacteria, while those fed with the salmon diet had developed a unique microbiota from the convergence of the diet, donor, and recipient microbiota. These results suggested that an intestinal microbiota transplantation may be effective if the basal microbiota from the gut is first cleared and a targeted dietary modification is provided to maintain and enrich the novel bacteria species over time.
Collapse
Affiliation(s)
- Alberto Ruiz
- Aquaculture Program, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Centre de La Ràpita, Crta. Poble Nou, km 5.5, 43540, La Ràpita, Spain.
| | - Enric Gisbert
- Aquaculture Program, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Centre de La Ràpita, Crta. Poble Nou, km 5.5, 43540, La Ràpita, Spain
| | - Karl B Andree
- Aquaculture Program, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Centre de La Ràpita, Crta. Poble Nou, km 5.5, 43540, La Ràpita, Spain
| |
Collapse
|
4
|
Yu A, Jansen MAC, Dalmeijer GW, Bruijning-Verhagen P, van der Ent CK, Grobbee DE, Burgner DP, Uiterwaal CSPM. Childhood infection burden, recent antibiotic exposure and vascular phenotypes in preschool children. PLoS One 2023; 18:e0290633. [PMID: 37713433 PMCID: PMC10503770 DOI: 10.1371/journal.pone.0290633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 08/11/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Severe childhood infection has a dose-dependent association with adult cardiovascular events and with adverse cardiometabolic phenotypes. The relationship between cardiovascular outcomes and less severe childhood infections is unclear. AIM To investigate the relationship between common, non-hospitalised infections, antibiotic exposure, and preclinical vascular phenotypes in young children. DESIGN A Dutch prospective population-derived birth cohort study. METHODS Participants were from the Wheezing-Illnesses-Study-Leidsche-Rijn (WHISTLER) birth cohort. We collected data from birth to 5 years on antibiotic prescriptions, general practitioner (GP)-diagnosed infections, and monthly parent-reported febrile illnesses (0-1 years). At 5 years, carotid intima-media thickness (CIMT), carotid artery distensibility, and blood pressure (BP) were measured. General linear regression models were adjusted for age, sex, smoke exposure, birth weight z-score, body mass index, and socioeconomic status. RESULTS Recent antibiotic exposure was associated with adverse cardiovascular phenotypes; each antibiotic prescription in the 3 and 6 months prior to vascular assessment was associated with an 18.1 μm (95% confidence interval, 4.5-31.6, p = 0.01) and 10.7 μm (0.8-20.5, p = 0.03) increase in CIMT, respectively. Each additional antibiotic prescription in the preceding 6 months was associated with an 8.3 mPa-1 decrease in carotid distensibility (-15.6- -1.1, p = 0.02). Any parent-reported febrile episode (compared to none) showed weak evidence of association with diastolic BP (1.6 mmHg increase, 0.04-3.1, p = 0.04). GP-diagnosed infections were not associated with vascular phenotypes. CONCLUSIONS Recent antibiotics are associated with adverse vascular phenotypes in early childhood. Mechanistic studies may differentiate antibiotic-related from infection-related effects and inform preventative strategies.
Collapse
Affiliation(s)
- Angela Yu
- Department of Paediatrics, Monash University, Clayton, Australia
| | - Maria A. C. Jansen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Geertje W. Dalmeijer
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Patricia Bruijning-Verhagen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Cornelis K. van der Ent
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Diederick E. Grobbee
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - David P. Burgner
- Department of Paediatrics, Monash University, Clayton, Australia
- Murdoch Children’s Research Institute, Parkville, Australia
- Department of Paediatrics, Melbourne University, Parkville, Australia
| | - Cuno S. P. M. Uiterwaal
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
5
|
Santana AB, Souto BS, Santos NCDM, Pereira JA, Tagliati CA, Novaes RD, Corsetti PP, de Almeida LA. Murine response to the opportunistic bacterium Pseudomonas aeruginosa infection in gut dysbiosis caused by 5-fluorouracil chemotherapy-induced mucositis. Life Sci 2022; 307:120890. [PMID: 35988752 DOI: 10.1016/j.lfs.2022.120890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/29/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
Abstract
AIMS This manuscript aims to explain the relationship between mucositis caused by 5-FU over gut bacterial species and susceptibility to opportunistic infection caused by P. aeruginosa. MAIN METHODS BALB/c mice were intraperitoneally treated with PBS or 5-FU. Bodyweight and faecal consistency were checked daily. Mice faecal DNA was extracted, and bacterial phylogenetic groups were analysed using qPCR or high-throughput sequencing. Immunofluorescence was used to evaluate BMDM activation by mice-treated faecal content. Mice were challenged intratracheally with virulent P. aeruginosa, and the CFU and histology were analysed. Faecal microbiota were transplanted to evaluate the gut microbiota and resistance to pulmonary P. aeruginosa recovery. KEY FINDINGS The animals treated with 5-FU presented mucositis with great weight loss, altered faecal consistency, bacterial gut dysbiosis and histological changes in the intestinal mucosa. Mice under 5-FU treatment were more susceptible to lung infection by the bacteria P. aeruginosa and had more extensive tissue damage during their lung infection with greater pro-inflammatory gene expression. It was observed that the mucositis remained in the groups with 5-FU even with the FMT. The results caused by mucositis in animals that received allogeneic FMT were reversed, however, with a decrease in P. aeruginosa susceptibility in animals treated with 5-FU and allogeneic FMT compared to animals treated with 5-FU and autologous FMT. SIGNIFICANCE Treatment with 5-FU in a murine model makes it more susceptible to pulmonary infection by the bacterium P. aeruginosa, FMT offers an opportunity to protect against this susceptibility to infection.
Collapse
Affiliation(s)
- Aleksander Brandão Santana
- Department of Microbiology and Immunology, Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas, Alfenas, Brazil
| | - Bianca Silva Souto
- Department of Microbiology and Immunology, Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas, Alfenas, Brazil
| | - Natália Cristina de Melo Santos
- Department of Microbiology and Immunology, Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas, Alfenas, Brazil
| | - Jéssica Assis Pereira
- Department of Microbiology and Immunology, Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas, Alfenas, Brazil
| | - Carlos Alberto Tagliati
- Laboratory of Toxicology (LabTox), Department of Clinical and Toxicological Analysis, Pharmacy Faculty, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rômulo Dias Novaes
- Department of Structural Biology, Federal University of Alfenas, Alfenas, Brazil
| | - Patrícia Paiva Corsetti
- Department of Microbiology and Immunology, Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas, Alfenas, Brazil
| | - Leonardo Augusto de Almeida
- Department of Microbiology and Immunology, Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas, Alfenas, Brazil.
| |
Collapse
|
6
|
Gobbo MM, Bomfim MB, Alves WY, Oliveira KC, Corsetti PP, de Almeida LA. Antibiotic-induced gut dysbiosis and autoimmune disease: A systematic review of preclinical studies. Clin Exp Rheumatol 2022; 21:103140. [PMID: 35830954 DOI: 10.1016/j.autrev.2022.103140] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/07/2022] [Indexed: 12/20/2022]
Abstract
Antibiotic-induced gut dysbiosis is believed to be associated with the onset and development of autoimmune diseases. To evaluate microbiota's variations triggered by antibiotic therapy and its outcomes on autoimmune diseases, preclinical studies regarding these subjects were included in this review. The studies were selected on PubMed, Scopus and Web of Science from 2011 to 2021 by three researchers that extracted study data and risk of bias, which were verified by a further 3 independent researchers. The team assessed the strength of evidence across studies. Of the eligible studies, 17 showed an improvement of the studied disease after antibiotic therapy and 10 had a negative effect on the course of the condition. The ameliorating factors of the studied diseases were mostly seen when using an antibiotic cocktail. Male animals had a good outcome after therapy and, for all genders, the increase in IL-10 and Treg cells was often shown to ameliorate disease after the antibiotic intervention. Firmicutes, Proteobacteria and Bacteroidetes appeared altered after the antibiotic intervention, leading to amelioration or worsening of the condition depending on the autoimmune disease. We identified that the number of autoimmune conditions approached leads to specific conclusions regarding the interventions, making it difficult to achieve an overall conclusion. Overall, even though pre-clinical studies must be translated to the human model, the studied aspects of gender, age, lineage and disease model substantially impact the outcomes that make for many intricacies that were not-established in the study of antibiotic-induced gut dysbiosis and autoimmunity.
Collapse
Affiliation(s)
- Marcela Mizuhira Gobbo
- Medical School, Federal University of Alfenas, Alfenas, Brazil; Department of Microbiology and Immunology, Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas, Alfenas, Brazil.
| | - Marina Bocamino Bomfim
- Medical School, Federal University of Alfenas, Alfenas, Brazil; Department of Microbiology and Immunology, Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas, Alfenas, Brazil
| | - Wille Ygor Alves
- Medical School, Federal University of Alfenas, Alfenas, Brazil; Department of Microbiology and Immunology, Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas, Alfenas, Brazil.
| | - Karen Cristina Oliveira
- Department of Microbiology and Immunology, Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas, Alfenas, Brazil
| | - Patrícia Paiva Corsetti
- Department of Microbiology and Immunology, Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas, Alfenas, Brazil
| | - Leonardo Augusto de Almeida
- Medical School, Federal University of Alfenas, Alfenas, Brazil; Department of Microbiology and Immunology, Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas, Alfenas, Brazil.
| |
Collapse
|
7
|
Exploring the Ecological Effects of Naturally Antibiotic-Insensitive Bifidobacteria in the Recovery of the Resilience of the Gut Microbiota during and after Antibiotic Treatment. Appl Environ Microbiol 2022; 88:e0052222. [PMID: 35652662 DOI: 10.1128/aem.00522-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Amoxicillin-clavulanic acid (AMC) is the most widely used antibiotic, being frequently prescribed to infants. Particular members of the genus Bifidobacterium are among the first microbial colonizers of the infant gut, and it has been demonstrated that they exhibit various activities beneficial for their human host, including promotion/maintenance of the human gut microbiota homeostasis. It has been shown that natural resistance of bifidobacteria to AMC is limited to a small number of strains. In the current study, we investigated the mitigation effects of AMC-resistant bifidobacteria in diversity preservation of the gut microbiota during AMC treatment. To this end, an in vitro coculture experiment based on infant fecal samples and an in vivo study employing a rodent model were performed. The results confirmed the ability of AMC-resistant bifidobacterial strains to bolster gut microbiota resilience, while specific covariance analysis revealed strain-specific and variable impacts on the microbiota composition by individual bifidobacterial taxa. IMPORTANCE The first microbial colonizers of the infant gut are members of the genus Bifidobacterium, which exhibit different activities beneficial to their host. Amoxicillin-clavulanic acid (AMC) is the most frequently prescribed antibiotic during infancy, and few strains of bifidobacteria are known to show a natural resistance to this antibiotic. In the present work, we evaluated the possible positive effects of AMC-resistant bifidobacterial strains in maintaining gut microbiota diversity during AMC exposure, performing an in vitro and in vivo experiment based on an infant gut model and a rodent model, respectively. Our results suggested the ability of AMC-resistant bifidobacterial strains to support gut microbiota restoration.
Collapse
|
8
|
Oral antibiotics reduce voluntary exercise behavior in athletic mice. Behav Processes 2022; 199:104650. [PMID: 35504410 DOI: 10.1016/j.beproc.2022.104650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022]
|
9
|
Bostanghadiri N, Ziaeefar P, Sameni F, Mahmoudi M, Hashemi A, Darban-Sarokhalil D. The controversial association of gut and urinary microbiota with kidney stone formation. Microb Pathog 2021; 161:105257. [PMID: 34687841 DOI: 10.1016/j.micpath.2021.105257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 09/24/2021] [Accepted: 10/15/2021] [Indexed: 12/17/2022]
Abstract
Nephrolithiasis (kidney stones) is one of the most common chronic kidney diseases that are typically more common among adult men comparing to adult women. The prevalence of this disease is increasing which is influenced by genetic and environmental factors. Kidney stones are mainly composed of calcium oxalate and urinary oxalate which is considered a dangerous factor in their formation. Besides diverse leading reasons in the progression of nephrolithiasis, the gut and urinary microbiome has been recognized as a major player in the development or prevention of it. These microbes produce metabolites that have diverse effects on host biological functions. Therefore, Changes in the composition and structure of the microbiome (dysbiosis) have been implicated in various diseases. The present review focuses on the roles of gut and urinary in kidney stone formation.
Collapse
Affiliation(s)
- Narjess Bostanghadiri
- -Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Pardis Ziaeefar
- -School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sameni
- -Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Mohammad Mahmoudi
- -Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- -Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Davood Darban-Sarokhalil
- -Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Tong Q, Cui LY, Bie J, Han XY, Hu ZF, Wang HB, Zhang JT. Changes in the gut microbiota diversity of brown frogs (Rana dybowskii) after an antibiotic bath. BMC Vet Res 2021; 17:333. [PMID: 34674716 PMCID: PMC8529755 DOI: 10.1186/s12917-021-03044-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 10/04/2021] [Indexed: 12/17/2022] Open
Abstract
Background Captive amphibians frequently receive antibiotic baths to control bacterial diseases. The potential collateral effect of these antibiotics on the microbiota of frogs is largely unknown. To date, studies have mainly relied on oral administration to examine the effects of antibiotics on the gut microbiota; in contrast, little is known regarding the effects of bath-applied antibiotics on the gut microbiota. The gut microbiota compositions of the gentamicin, recovery, and control groups were compared by Illumina high-throughput sequencing, and the functional profiles were analysed using Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt). Furthermore, the relationship between the structure and predicted functional composition of the gut microbiota was determined. Results The alpha diversity indices were significantly reduced by the gentamicin bath, illustrating that this treatment significantly changed the composition of the gut microbiota. After 7 days, the gut microbiota of the recovery group was not significantly different from that of the gentamicin group. Forty-four indicator taxa were selected at the genus level, comprising 42 indicators representing the control group and 2 indicators representing the gentamicin and recovery groups. Potential pathogenic bacteria of the genera Aeromonas, Citrobacter, and Chryseobacterium were significantly depleted after the gentamicin bath. There was no significant positive association between the community composition and functional composition of the gut microbiota in the gentamicin or control frogs, indicating that the functional redundancy of the gut bacterial community was high. Conclusions Gentamicin significantly changed the structure of the gut microbiota of R. dybowskii, and the gut microbiota exhibited weak resilience. However, the gentamicin bath did not change the functional composition of the gut microbiota of R. dybowskii, and there was no significant correlation between the structural composition and the functional composition of the gut microbiota. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-021-03044-z.
Collapse
Affiliation(s)
- Qing Tong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.,Jiamusi Branch of Heilongjiang Academy of Forestry Sciences, Jiamusi, 154002, China.,College of Life Science, Jiamusi University, Jiamusi, 154007, China
| | - Li-Yong Cui
- Jiamusi Branch of Heilongjiang Academy of Forestry Sciences, Jiamusi, 154002, China
| | - Jia Bie
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xiao-Yun Han
- College of Life Science, Jiamusi University, Jiamusi, 154007, China
| | - Zong-Fu Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Hong-Bin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jian-Tao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
11
|
Freccero F, Lanci A, Mariella J, Viciani E, Quercia S, Castagnetti A, Castagnetti C. Changes in the Fecal Microbiota Associated with a Broad-Spectrum Antimicrobial Administration in Hospitalized Neonatal Foals with Probiotics Supplementation. Animals (Basel) 2021; 11:ani11082283. [PMID: 34438741 PMCID: PMC8388449 DOI: 10.3390/ani11082283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Post-antibiotic intestinal dysbiosis leads to an overall reduction in bacterial and functional diversity, along with a minor resistance against pathogens. The study aimed to determine the changes on the fecal microbiota in hospitalized neonatal foals administered with broad-spectrum antimicrobials and supplemented probiotics. Fecal samples were collected at hospital admission, at the end of the antimicrobial treatment and at discharge. Seven foals treated with intravenous ampicillin and aminoglycosides for a mean of seven days were included. The results suggest that the fecal microbiota of neonatal foals rapidly returns to a high diversity after treatment. While the findings need to be confirmed in a larger population, the study suggests that in foals, the effect of antimicrobials may be strongly influenced by the changes that occur over time in the developing gut microbiota. Of note, the findings are influenced by the use of probiotics, and whether the changes would be consistent in antimicrobial-administered but not supplemented foals remains to be elucidated. Abstract There is a wide array of evidence across species that exposure to antibiotics is associated with dysbiosis, and due to their widespread use, this also raises concerns also in medicine. The study aimed to determine the changes on the fecal microbiota in hospitalized neonatal foals administered with broad-spectrum antimicrobials and supplemented probiotics. Fecal samples were collected at hospital admission (Ta), at the end of the antimicrobial treatment (Te) and at discharge (Td). Feces were analysed by next-generation sequencing of the 16S rRNA gene on Illumina MiSeq. Seven foals treated with IV ampicillin and amikacin/gentamicin were included. The mean age at Ta was 19 h, the mean treatment length was 7 days and the mean time between Te and Td was 4.3 days. Seven phyla were identified: Actinobacteria, Bacteroidetes, Firmicutes, Fusobacteria, Proteobacteria, TM7 and Verrucomicrobia. At Ta, Firmicutes (48.19%) and Proteobacteria (31.56%) were dominant. The alpha diversity decreased from Ta to Te, but it was the highest at Td. The beta diversity was higher at Ta than at Te and higher at Td than at Te. An increase in Akkermansia over time was detected. The results suggest that the intestinal microbiota of neonatal foals rapidly returns to a high diversity after treatment. It is possible that in foals, the effect of antimicrobials is strongly influenced or overshadowed by the time-dependent changes in the developing gut microbiota.
Collapse
Affiliation(s)
- Francesca Freccero
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano Dell’Emilia, 40064 Bologna, Italy; (F.F.); (J.M.); (C.C.)
| | - Aliai Lanci
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano Dell’Emilia, 40064 Bologna, Italy; (F.F.); (J.M.); (C.C.)
- Correspondence:
| | - Jole Mariella
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano Dell’Emilia, 40064 Bologna, Italy; (F.F.); (J.M.); (C.C.)
| | - Elisa Viciani
- Wellmicro srl, Via Piero Gobetti 101, 40127 Bologna, Italy; (E.V.); (S.Q.); (A.C.)
| | - Sara Quercia
- Wellmicro srl, Via Piero Gobetti 101, 40127 Bologna, Italy; (E.V.); (S.Q.); (A.C.)
| | - Andrea Castagnetti
- Wellmicro srl, Via Piero Gobetti 101, 40127 Bologna, Italy; (E.V.); (S.Q.); (A.C.)
| | - Carolina Castagnetti
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano Dell’Emilia, 40064 Bologna, Italy; (F.F.); (J.M.); (C.C.)
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Via Tolara di Sopra 41/E, Ozzano dell’Emilia, 40064 Bologna, Italy
| |
Collapse
|
12
|
Ghanem S, Kim CJ, Dutta D, Salifu M, Lim SH. Antimicrobial therapy during cancer treatment: Beyond antibacterial effects. J Intern Med 2021; 290:40-56. [PMID: 33372309 DOI: 10.1111/joim.13238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/22/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023]
Abstract
Cancer treatment options have evolved to include immunotherapy and targeted therapy, in addition to traditional chemoradiation. Chemoradiation places the patient at a higher risk of infection through a myelosuppressive effect. High clinical suspicion and early use of antimicrobials play a major role in decreasing any associated morbidity and mortality. This has led to a widespread use of antimicrobials in cancer patients. Antimicrobial use, however, does not come without its perils. Dysbiosis caused by antimicrobial use affects responses to chemotherapeutic agents and is prognostic in the development and severity of certain cancer treatment-related complications such as graft-versus-host disease and Clostridioides difficile infections. Studies have also demonstrated that an intact gut microbiota is essential in the anticancer immune response. Antimicrobial use can therefore modulate responses and outcomes with immunotherapy targeting immune checkpoints. In this review, we highlight the perils associated with antimicrobial use during cancer therapy and the importance of a more judicious approach. We discuss the nature of the pathologic changes in the gut microbiota resulting from antimicrobial use. We explore the effect these changes have on responses and outcomes to different cancer treatment modalities including chemotherapy and immunotherapy, as well as potential adverse clinical consequences in the setting of stem cell transplant.
Collapse
Affiliation(s)
- S Ghanem
- From the, Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - C J Kim
- From the, Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - D Dutta
- From the, Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - M Salifu
- From the, Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - S H Lim
- From the, Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| |
Collapse
|
13
|
Ray P, Pandey U, Aich P. Comparative analysis of beneficial effects of vancomycin treatment on Th1- and Th2-biased mice and the role of gut microbiota. J Appl Microbiol 2020; 130:1337-1356. [PMID: 32955795 DOI: 10.1111/jam.14853] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 08/18/2020] [Accepted: 09/07/2020] [Indexed: 12/22/2022]
Abstract
AIMS The aim was to understand the time-dependent antibiotic-induced perturbation pattern of gut microbiota and its effect on the innate immune and metabolic profile of the host. METHODS AND RESULTS Vancomycin was administered at 50 mg kg-1 of body weight twice daily for six consecutive days to perturb the gut microbiota of C57BL/6 (Th1-biased) and BALB/c (Th2-biased) mice. Following treatment with vancomycin, we observed a reduction in the abundance of phyla Firmicutes and Bacteroides and an increase in Proteobacteria in the gut for both strains of mice following treatment with vancomycin till day 4. Abundance of Akkermansia muciniphila of Verrucomicrobia phylum also increased, from day 5 onwards following vancomycin treatment. The time-dependent variation of gut microbiota was associated with increased (i) expression of toll-like receptors and inflammatory genes such as TNF-α, IL-6, and IL-17, (ii) gut barrier permeability and (iii) blood glucose level of the host. The results also showed that (i) transplantation of cecal microbiota from vancomycin-treated day 6 mice to day 3 vancomycin-treated mice helped in restoring blood glucose level in C57BL/6 mice and (ii) short-chain fatty acids like acetate, butyrate and propionate changed with the alteration of gut microbiota to induce differential regulation of host immune response. CONCLUSIONS The current results revealed that an increase in A. muciniphila led to decreased inflammation and increased rate of glucose tolerance in the host. The treatment, with vancomycin till day 4, increased expression of inflammatory genes. The continuation of vancomycin for two more days reversed the effects. The effects were significantly more in C57BL/6 than BALB/c mice. SIGNIFICANCE AND IMPACT OF THE STUDY The current study established that the treatment with vancomycin till day 4 increased pathogenic bacteria but day 5 onwards provided significant health-related benefits to the host by increasing A. muciniphila more in C57BL/6 than BALB/c mice.
Collapse
Affiliation(s)
- P Ray
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, Khurdha, Odisha, India
| | - U Pandey
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, Khurdha, Odisha, India
| | - P Aich
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, Khurdha, Odisha, India
| |
Collapse
|
14
|
Абатуров А, Никулина А. Antibiotic Therapy as a Risk Factor of Obesity Development in Children. ПЕДИАТРИЯ. ВОСТОЧНАЯ ЕВРОПА 2020:268-290. [DOI: 10.34883/pi.2020.8.2.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Настоящий обзор научной литературы посвящен вопросам, связанным с механизмами антибактериально-индуцированного адипогенеза. Антибиотиками, наиболее высоко ассоциированными с развитием ожирения у детей, считают: амоксициллин, цефотаксим, макролиды, тетрациклины, ванкомицин. На основании результатов филогенетических, метагеномных исследований эффектов антибиотиков установлено, что их применение в антенатальном, раннем постнатальном периоде приводит к пролонгированным изменениям как состава, так и функционирования микробиома, которые ассоциированы с повышенным риском последующего увеличения массы тела ребенка. Механизмы непосредственного влияния антибиотиков на адипогенез связаны с их способностью повышать аппетит за счет стимуляции высвобождения орексина и меланин-концентрирующего гормона; увеличивать абсорбцию пищевых ингредиентов; активировать липогенез; индуцировать митохондриальную дисфункцию и тем самым способствовать накоплению жирных кислот. Применение антибиотиков существенно изменяет структуру микробиома кишечника, а именно: развитие ожирения связано с высоким уровнем представительства бактерий филюмов Actinobacteria и Firmicutes в сочетании со снижением численности бактерий Bacteroidetes, Verrucomicrobia и Faecalibacterium prausnitzii. Антибиотик-индуцированные изменения микробиома могут существенно влиять на аппетит, так как уровень грелина, вызывающего аппетит, положительно коррелирует с представительством бактерий Bacteroides и Prevotella, и отрицательно – с численностью бактерий Bifidobacterium, Lactobacillus, Blautia coccoides и Eubacterium rectale. Доказано, что применение некоторых антибиотиков сопровождается не только накоплением висцерального жира, но и приводит к развитию как неалкогольной болезни печени, так и инсулинорезистентности. Рецепторы FXR и TGR5 являются сенсорами изменений микробиоты кишечника, которые участвуют в регуляции метаболических процессов макроорганизма. Развитие ожирения характеризуется наличием низкоуровневого системного воспаления. При развитии ожирения по мере увеличения размеров адипоцитов фенотип макрофагов меняется на провоспалительный фенотип М1. Накопление провоспалительных клеток в висцеральной жировой ткани является важной причиной развития инсулинорезистентности. В настоящее время необходимость применения антибиотиков при лечении инфекционных заболеваний, вызванных бактериальными агентами, не вызывает никаких клинических сомнений. Однако появление научных сведений о метаболических эффектах, возникновение которых ассоциировано с антибиотикотерапией, ставит клинические новые задачи, решение которых, вероятно, лежит в оптимизации режимов применения антибиотиков и выборе сопровождающих лекарственных средств.
This review of scientific literature is devoted to issues related to the mechanisms of antibacterial- induced adipogenesis. The antibiotics most highly associated with the development of obesity in children are the following: amoxicillin, cefotaxime, macrolides, tetracyclines, vancomycin. On the base of the results of phylogenetic, metagenomic studies of the effects of antibiotics, it was found that their use in the antenatal, early postnatal period leads to prolonged changes in both the composition and functioning of the microbiome, which is associated with the increased risk of subsequent increase of body weight of the child. The mechanisms of direct effect of antibiotics on adipogenesis are associated with their ability to increase appetite, by stimulating the release of orexin and melanin-concentrating hormone; increase the absorption of food ingredients; activate lipogenesis; induce mitochondrial dysfunction and thereby contribute to accumulation of fatty acids. The use of antibiotics significantly changes the structure of the intestinal microbiome, namely, the development of obesity is associated with a high representation of phylum bacteria Actinobacteria and Firmicutes in combination with the decrease of the number of bacteria Bacteroidetes, Verrucomicrobia and Faecalibacterium prausnitzii. Antibiotic-induced changes in the microbiome can significantly affect appetite, because the level of ghrelin that causes appetite positively correlates with the presence of bacteria Bacteroides and Prevotella, and negatively with the number of bacteria Bifidobacterium, Lactobacillus, Blautia coccoides and Eubacterium rectale. It was proved that the use of certain antibiotics is accompanied not only by the accumulation of visceral fat, but also leads to the development of both non-alcoholic liver disease and insulin resistance. The FXR and TGR5 receptors are the sensors of changes in the intestinal microbiota, which is involved in the regulation of the metabolic processes of the macroorganism. The development of obesity is characterized by the presence of low-level systemic inflammation. With the development of obesity, as the size of adipocytes increases, the macrophage phenotype changes to the pro- inflammatory M1 phenotype. The accumulation of pro-inflammatory cells in visceral adipose tissue is an important reason for development of insulin resistance. Currently, the need for antibiotics in the treatment of infectious diseases caused by bacterial agents does not raise any clinical doubts. However, the emergence of scientific information about metabolic effects, the occurrence of which is associated with antibiotic therapy, presents new clinical challenges, the solution of which probably lies in optimizing antibiotic regimens and choosing the accompanying drugs.
Collapse
|
15
|
Johnson KVA, Burnet PWJ. Opposing effects of antibiotics and germ-free status on neuropeptide systems involved in social behaviour and pain regulation. BMC Neurosci 2020; 21:32. [PMID: 32698770 PMCID: PMC7374917 DOI: 10.1186/s12868-020-00583-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 07/07/2020] [Indexed: 12/16/2022] Open
Abstract
Background Recent research has revealed that the community of microorganisms inhabiting the gut affects brain development, function and behaviour. In particular, disruption of the gut microbiome during critical developmental windows can have lasting effects on host physiology. Both antibiotic exposure and germ-free conditions impact the central nervous system and can alter multiple aspects of behaviour. Social impairments are typically displayed by antibiotic-treated and germ-free animals, yet there is a lack of understanding of the underlying neurobiological changes. Since the μ-opioid, oxytocin and vasopressin systems are key modulators of mammalian social behaviour, here we investigate the effect of experimentally manipulating the gut microbiome on the expression of these pathways. Results We show that social neuropeptide signalling is disrupted in germ-free and antibiotic-treated mice, which may contribute to the behavioural deficits observed in these animal models. The most notable finding is the reduction in neuroreceptor gene expression in the frontal cortex of mice administered an antibiotic cocktail post-weaning. Additionally, the changes observed in germ-free mice were generally in the opposite direction to the antibiotic-treated mice. Conclusions Antibiotic treatment when young can impact brain signalling pathways underpinning social behaviour and pain regulation. Since antibiotic administration is common in childhood and adolescence, our findings highlight the potential adverse effects that antibiotic exposure during these key neurodevelopmental periods may have on the human brain, including the possible increased risk of neuropsychiatric conditions later in life. In addition, since antibiotics are often considered a more amenable alternative to germ-free conditions, our contrasting results for these two treatments suggest that they should be viewed as distinct models.
Collapse
Affiliation(s)
- Katerina V A Johnson
- Department of Experimental Psychology, University of Oxford, Radcliffe Observatory Quarter, Oxford, OX2 6GG, UK. .,Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX, UK.
| | - Philip W J Burnet
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX, UK
| |
Collapse
|
16
|
Marques AM, Sarandy MM, Novaes RD, Gonçalves RV, Freitas MB. Preclinical relevance of probiotics in type 2 diabetes: A systematic review. Int J Exp Pathol 2020; 101:68-79. [PMID: 32608551 DOI: 10.1111/iep.12359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 05/20/2020] [Accepted: 05/24/2020] [Indexed: 12/14/2022] Open
Abstract
Type 2 diabetes (T2DM) is among the most prevalent metabolic diseases in the world and may result in several long-term complications. The crosstalk between gut microbiota and host metabolism is closely related to T2DM. Currently, fragmented data hamper defining the relationship between probiotics and T2DM. This systematic review aimed at investigating the effects of probiotics on T2DM in animal models. We systematically reviewed preclinical evidences using PubMed/MEDLINE and Scopus databases, recovering 24 original articles published until September 27th, 2019. This systematic review was performed according to PRISMA guidelines. We included experimental studies with animal models reporting the effects of probiotics on T2DM. Studies were sorted by characteristics of publications, animal models, performed analyses, probiotic used and interventions. Bias analysis and methodological quality assessments were examined through the SYRCLE's Risk of Bias tool. Probiotics improved T2DM in 96% of the studies. Most studies (96%) used Lactobacillus strains, and all of them led to improved glycaemia. All studies used rodents as models, and male animals were preferred over females. Results suggest that probiotics have a beneficial effect in T2DM animals and could be used as a supporting alternative in the disease treatment. Considering a detailed evaluation of the reporting and methodological quality, the current preclinical evidence is at high risk of bias. We hope that our critical analysis will be useful in mitigating the sources of bias in further studies.
Collapse
Affiliation(s)
| | | | - Rômulo Dias Novaes
- Department of Structural Biology, Federal University of Alfenas, Alfenas, Brazil
| | | | | |
Collapse
|
17
|
Silva EN, Martins TVF, Miyauchi-Tavares TM, Miranda BAE, Dos Santos GDA, Rosa CP, Santos JA, Novaes RD, de Almeida LA, Corsetti PP. Amoxicillin-induced gut dysbiosis influences estrous cycle in mice and cytokine expression in the ovary and the caecum. Am J Reprod Immunol 2020; 84:e13247. [PMID: 32304259 DOI: 10.1111/aji.13247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/03/2020] [Accepted: 04/09/2020] [Indexed: 12/22/2022] Open
Abstract
PROBLEM Gut dysbiosis is caused by several factors, including the use of antibiotics. Since intestinal dysbiosis is associated with a wide range of immunopathological and reproductive conditions, the main goal of this study was to evaluate amoxicillin-induced gut dysbiosis and its influence on the oestrous cycle in mice. METHOD OF STUDY Mice were treated with amoxicillin or PBS, and faecal microbiota was evaluated by 16S rDNA metagenomic sequencing. The oestrous cycle was evaluated by vaginal cytology, vaginal opening and flow cytometry. After the induction of gut dysbiosis, the ovaries and the caecum were analysed to differential expression of IL-1β and IL-10 genes and histological analysis. RESULTS Amoxicillin-treated mice presented differing bacterial groups in the faecal microbiota when compared to the PBS-treated group indicating that amoxicillin treatment-induced gut dysbiosis and they gained weight. The vaginal cytology analysis showed that amoxicillin-induced gut dysbiosis decreased the number of cells but increased the relative number of leucocytes and altered the oestrous cycle. IL-1β was shown to be upregulated in the caecum and in the ovary of the dysbiotic mice. On the other hand, IL-10 expression was shown to be diminished in both organs of the dysbiotic mice. The oocyte area from dysbiotic group presented lower than non-dysbiotic mice with increasing thickness of the pellucid zone. The follicular teak from dysbiotic mice showed lower thickness than non-dysbiotic mice. CONCLUSION The results indicate that amoxicillin induces gut dysbiosis and influences the oestrous cycle and the inflammatory status of the ovary and the caecum.
Collapse
Affiliation(s)
- Evandro Neves Silva
- Laboratório de Imunologia das Doenças Infecciosas e Crônicas, Universidade José do Rosário Vellano, Alfenas, Brazil
| | - Thaís Viana Fialho Martins
- Laboratório de Imunologia das Doenças Infecciosas e Crônicas, Universidade José do Rosário Vellano, Alfenas, Brazil
| | | | | | - Gabriela de Assis Dos Santos
- Laboratório de Imunologia das Doenças Infecciosas e Crônicas, Universidade José do Rosário Vellano, Alfenas, Brazil
| | - Caio Pupin Rosa
- Departamento de Microbiologia e Imunologia, Universidade Federal de Alfenas, Alfenas, Brazil
| | - Jeferson Antônio Santos
- Laboratório de Imunologia das Doenças Infecciosas e Crônicas, Universidade José do Rosário Vellano, Alfenas, Brazil
| | - Rômulo Dias Novaes
- Departamento de Biologia Estrutural, Universidade Federal de Alfenas, Alfenas, Brazil
| | | | - Patrícia Paiva Corsetti
- Laboratório de Imunologia das Doenças Infecciosas e Crônicas, Universidade José do Rosário Vellano, Alfenas, Brazil
| |
Collapse
|
18
|
Payer M, Tan WC, Han J, Ivanovski S, Mattheos N, Pjetursson BE, Zhuang L, Fokas G, Wong MCM, Acham S, Lang NP, Hafner E, Jakse N, Kirnbauer B, Stagnell S, Simon S, Truschnegg A, Feng G, Meng X, Shi J. The effect of systemic antibiotics on clinical and patient‐reported outcome measures of oral implant therapy with simultaneous guided bone regeneration. Clin Oral Implants Res 2020; 31:442-451. [DOI: 10.1111/clr.13580] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 12/29/2019] [Accepted: 12/30/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Michael Payer
- Department of Oral Surgery and Orthodontics University Clinic of Dental Medicine & Oral Health Medical University Graz Graz Austria
| | - Wah Ching Tan
- National Dental Centre Singapore Singapore City Singapore
| | - Jie Han
- School of Stomatology Peking University Beijing China
| | - Saso Ivanovski
- School of Dentistry and Oral Health Griffith University Gold Coast QLD Australia
| | - Nikos Mattheos
- Faculty of Dentistry The University of Hong Kong Hong Kong SAR China
| | | | - Longfei Zhuang
- Department of Implant Dentistry Shanghai Ninth People's Hospital Shanghai Jiao Tong University Shanghai China
| | - George Fokas
- Faculty of Dentistry The University of Hong Kong Hong Kong SAR China
| | - May C. M. Wong
- Faculty of Dentistry The University of Hong Kong Hong Kong SAR China
| | - Stephan Acham
- Department of Oral Surgery and Orthodontics University Clinic of Dental Medicine & Oral Health Medical University Graz Graz Austria
| | - Niklaus P. Lang
- Faculty of Dentistry The University of Hong Kong Hong Kong SAR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Savcı A, Koçpınar EF, Budak H, Çiftci M, Şişecioğlu M. The Effects of Amoxicillin, Cefazolin, and Gentamicin Antibiotics on the Antioxidant System in Mouse Heart Tissues. Protein Pept Lett 2020; 27:614-622. [PMID: 31721686 DOI: 10.2174/0929866526666191112125949] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/27/2019] [Accepted: 09/18/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Free radicals lead to destruction in various organs of the organism. The improper use of antibiotics increases the formation of free radicals and causes oxidative stress. OBJECTIVE In this study, it was aimed to determine the effects of gentamicin, amoxicillin, and cefazolin antibiotics on the mouse heart. METHODS 20 male mice were divided into 4 groups (1st control, 2nd amoxicillin, 3rd cefazolin, and 4th gentamicin groups). The mice in the experimental groups were administered antibiotics intraperitoneally at a dose of 100 mg / kg for 6 days. The control group received normal saline in the same way. The gene expression levels and enzyme activities of SOD, CAT, GPx, GR, GST, and G6PD antioxidant enzymes were investigated. RESULTS GSH levels decreased in both the amoxicillin and cefazolin groups, while GR, CAT, and SOD enzyme activities increased. In the amoxicillin group, Gr, Gst, Cat, and Sod gene expression levels increased. CONCLUSION As a result, it was concluded that amoxicillin and cefazolin caused oxidative stress in the heart, however, gentamicin did not cause any effects.
Collapse
Affiliation(s)
- Ahmet Savcı
- Department of Chemistry, Faculty of Art and Science, Bingol University, Bingol, Turkey
| | - Enver Fehim Koçpınar
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Mus Alparslan University, Mus, Turkey
| | - Harun Budak
- Department of Molecular Biology and Genetics, Faculty of Science, Ataturk University, Erzurum, Turkey
| | - Mehmet Çiftci
- Department of Chemistry, Faculty of Art and Science, Bingol University, Bingol, Turkey
| | - Melda Şişecioğlu
- Department of Molecular Biology and Genetics, Faculty of Science, Ataturk University, Erzurum, Turkey
| |
Collapse
|
20
|
Whey Protein Hydrolysate and Pumpkin Pectin as Nutraceutical and Prebiotic Components in a Functional Mousse with Antihypertensive and Bifidogenic Properties. Nutrients 2019; 11:nu11122930. [PMID: 31816861 PMCID: PMC6950020 DOI: 10.3390/nu11122930] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 02/06/2023] Open
Abstract
Systematical consumption of functional products has a significant positive effect on health and can reduce the risk of diseases. The aim of this study was to investigate the possibility of using whey protein hydrolysate (WPH) and pumpkin pectin as ingredients in a functional mousse, to evaluate the mousse’s antioxidant and hypotensive activities in vitro, and to evaluate the effect of the long-term intake of mousse samples on the progression of hypertension in spontaneously hypertensive rats (SHRs) and on the microbiome status in Wistar rats with antibiotic-induced dysbiosis. The experimental mousse’s in vitro antioxidant activity (oxygen radical absorbance capacity) increased by 1.2 times. The hypotensive (angiotensin-1-converting enzyme inhibitory) activity increased by 6 times in comparison with a commercial mousse. Moreover, the addition of pectin allowed the elimination of the bitter aftertaste of WPH. In vivo testing confirmed the hypotensive properties of the experimental mousse. The systolic blood pressure in SHRs decreased by 18 mmHg and diastolic blood pressure by 12 mmHg. The experimental mousse also showed a pronounced bifidogenic effect. The Bifidobacterium spp. population increased by 3.7 times in rats orally administered with the experimental mousse. The results of these studies confirm that WPH and pumpkin pectin are prospective ingredients for the development of functional mousses.
Collapse
|
21
|
Rosa CP, Pereira JA, Cristina de Melo Santos N, Brancaglion GA, Silva EN, Tagliati CA, Novaes RD, Corsetti PP, de Almeida LA. Vancomycin-induced gut dysbiosis during Pseudomonas aeruginosa pulmonary infection in a mice model. J Leukoc Biol 2019; 107:95-104. [PMID: 31682032 DOI: 10.1002/jlb.4ab0919-432r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 09/27/2019] [Accepted: 10/23/2019] [Indexed: 12/25/2022] Open
Abstract
Pseudomonas aeruginosa is one of the most common opportunistic pathogens causing respiratory infections in hospitals. Vancomycin, the antimicrobial agent usually used to treat bacterial nosocomial infections, is associated with gut dysbiosis. As a lung-gut immunologic axis has been described, this study aimed to evaluate both the immunologic and histopathologic effects on the lungs and the large intestine resulting from vancomycin-induced gut dysbiosis in the P. aeruginosa pneumonia murine model. Metagenomic analysis demonstrated that vancomycin-induced gut dysbiosis resulted in higher Proteobacteria and lower Bacteroidetes populations in feces. Given that gut dysbiosis could augment the proinflammatory status of the intestines leading to a variety of acute inflammatory diseases, bone marrow-derived macrophages were stimulated with cecal content from dysbiotic mice showing a higher expression of proinflammatory cytokines and lower expression of IL-10. Dysbiotic mice showed higher levels of viable bacteria in the lungs and spleen when acutely infected with P. aeruginosa, with more lung and cecal damage and increased IL-10 expression in bronchoalveolar lavage. The susceptible and tissue damage phenotype was reversed when dysbiotic mice received fecal microbiota transplantation. In spite of higher recruitment of CD11b+ cells in the lungs, there was no higher CD80+ expression, DC+ cell amounts or proinflammatory cytokine expression. Taken together, our results indicate that the bacterial community found in vancomycin-induced dysbiosis dysregulates the gut inflammatory status, influencing the lung-gut immunologic axis to favor increased opportunistic infections, for example, by P. aeruginosa.
Collapse
Affiliation(s)
- Caio Pupin Rosa
- Department of Microbiology and Immunology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Jéssica Assis Pereira
- Department of Microbiology and Immunology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | | | | | - Evandro Neves Silva
- Laboratory of Infectious and Chronic Diseases (LIDIC), José do Rosário Vellano University, Alfenas, Minas Gerais, Brazil
| | - Carlos Alberto Tagliati
- Laboratory of Toxicology (LabTox), Department of Clinical and Toxicological Analysis, Pharmacy Faculty, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rômulo Dias Novaes
- Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Patrícia Paiva Corsetti
- Laboratory of Infectious and Chronic Diseases (LIDIC), José do Rosário Vellano University, Alfenas, Minas Gerais, Brazil
| | | |
Collapse
|
22
|
Jing Y, Liu H, Xu W, Yang Q. 4,4′‐Diaponeurosporene‐ProducingBacillus subtilisPromotes the Development of the Mucosal Immune System of the Piglet Gut. Anat Rec (Hoboken) 2019; 302:1800-1807. [DOI: 10.1002/ar.24102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 11/06/2018] [Accepted: 11/21/2018] [Indexed: 01/14/2023]
Affiliation(s)
- Yuchao Jing
- College of Veterinary MedicineNanjing Agricultural University Nanjing Jiangsu 210095 People's Republic of China
| | - Haofei Liu
- College of Veterinary MedicineNanjing Agricultural University Nanjing Jiangsu 210095 People's Republic of China
| | - Wenwen Xu
- College of Veterinary MedicineNanjing Agricultural University Nanjing Jiangsu 210095 People's Republic of China
| | - Qian Yang
- College of Veterinary MedicineNanjing Agricultural University Nanjing Jiangsu 210095 People's Republic of China
| |
Collapse
|
23
|
Uremic Fetor Revisited: Exhaled Volatile Biomarkers in Acute Kidney Injury. Crit Care Med 2019; 47:299-300. [PMID: 30653063 DOI: 10.1097/ccm.0000000000003584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
24
|
Liang W, Huang Y, Tan X, Wu J, Duan J, Zhang H, Yin B, Li Y, Zheng P, Wei H, Xie P. Alterations Of Glycerophospholipid And Fatty Acyl Metabolism In Multiple Brain Regions Of Schizophrenia Microbiota Recipient Mice. Neuropsychiatr Dis Treat 2019; 15:3219-3229. [PMID: 31819450 PMCID: PMC6876209 DOI: 10.2147/ndt.s225982] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/11/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Schizophrenia is a debilitating psychiatric disorder characterized by molecular and anatomical abnormalities of multiple brain regions. Our recent study showed that dysbiosis of the gut microbiota contributes to the onset of schizophrenia-relevant behaviors, but the underlying mechanisms remain largely unknown. PURPOSE This study aimed to investigate how gut microbiota shapes metabolic signatures in multiple brain regions of schizophrenia microbiota recipient mice. METHODS Gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-mass spectrometry (LC-MS) were used to compare the metabolic signatures in the cortex, cerebellum and striatum of schizophrenia microbiota and healthy microbiota recipient mice. Enrichment analysis was further conducted to uncover the crucial metabolic pathways related to schizophrenia-relevant behaviors. RESULTS We found that the metabolic phenotypes of these three regions were substantially different in schizophrenia microbiota recipient mice from those in healthy microbiota recipient mice. In total, we identified 499 differential metabolites that could discriminate the two groups in the three brain regions. These differential metabolites were mainly involved in glycerophospholipid and fatty acyl metabolism. Moreover, we found four of fatty acyl metabolites that were consistently altered in the three brain regions. CONCLUSION Taken together, our study suggests that alterations of glycerophospholipid and fatty acyl metabolism are implicated in the onset of schizophrenia-relevant behaviors, which may provide a new understanding of the etiology of schizophrenia.
Collapse
Affiliation(s)
- Weiwei Liang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, People's Republic of China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yu Huang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xunmin Tan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jing Wu
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China.,The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jiajia Duan
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China.,The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Hanping Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Bangmin Yin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yifan Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Hong Wei
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China
| |
Collapse
|