1
|
Marquez-Paradas E, Torrecillas-Lopez M, Barrera-Chamorro L, del Rio-Vazquez JL, Gonzalez-de la Rosa T, Montserrat-de la Paz S. Microbiota-derived extracellular vesicles: current knowledge, gaps, and challenges in precision nutrition. Front Immunol 2025; 16:1514726. [PMID: 40051622 PMCID: PMC11882860 DOI: 10.3389/fimmu.2025.1514726] [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: 10/21/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
The gut microbiota has co-evolved with its host, profoundly shaping the development and functioning of the immune system. This co-evolution has led to a dynamic relationship where microbial metabolites and molecular signals influence immune maturation, tolerance, and defense mechanisms, highlighting its essential role in maintaining host health. Recently, bacterial extracellular vesicles (BEVs), membrane nanoparticles produced by bacteria, have emerged as important players in gut balance and as potent immune modulators. These vesicles reflect the characteristics of the bacterial membrane and contain nucleic acids, proteins, lipids, and metabolites. They can regulate immune processes and are involved in neurological and metabolic diseases due to their ability to distribute both locally in the gut and systemically, affecting immune responses at both levels. This review provides a comprehensive overview of the characteristics and functional profile of BEVs, detailing how nutrition influences the production and function of these vesicles, how antibiotics can disrupt or alter their composition, and how these factors collectively impact immunity and disease development. It also highlights the potential of BEVs in the development of precision nutritional strategies through dietary modulation, such as incorporating prebiotic fibers to enhance beneficial BEV production, reducing intake of processed foods that may promote harmful BEVs, and tailoring probiotic interventions to influence specific microbial communities and their vesicular outputs.
Collapse
Affiliation(s)
- Elvira Marquez-Paradas
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| | - Maria Torrecillas-Lopez
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| | - Luna Barrera-Chamorro
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| | - Jose L. del Rio-Vazquez
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Teresa Gonzalez-de la Rosa
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| | - Sergio Montserrat-de la Paz
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| |
Collapse
|
2
|
Gałęcka I, Rychlik A, Całka J. Influence of selected dosages of plastic microparticles on the porcine fecal microbiome. Sci Rep 2025; 15:1269. [PMID: 39779716 PMCID: PMC11711237 DOI: 10.1038/s41598-024-80337-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025] Open
Abstract
Studies conducted so far have shown that nano- and microplastic may disturb the intestinal microenvironment by interacting with the intestinal epithelium and the gut microbiota. Depending on the research model used, the effect on the microbiome is different-an increase or decrease in selected taxa resulting in the development of dysbiosis. Dysbiosis may be associated with intestinal inflammation, development of mental disorders or diabetes. The aim of the study was to analyze the intestinal microbiome in 15 gilts divided into 3 research groups (n = 5; control group, receiving micropartices at a dose 0.1 g/day (LD) and 1 g/day (HD)). Feaces were collected before and after 28 days of exposure to PET microplastics. The analysis of the intestinal microbiome was performed using next-generation sequencing. Alpha and beta diversity indices were compared, showing, that repetition affected only the abundance indices in the control and LD groups, but not in the HD group. The relationships between the number of reads at the phylum, genus and species level and the microplastic dose were calculated using statistical methods (r-Pearson correlation, generalized regression model, analysis of variance). The statistical analysis revealed, that populations of Family XIII AD3011 group, Coprococcus, V9D2013 group, UCG-010 and Sphaerochaeta increased with increasing MP-PET dose. The above-mentioned taxa are mainly responsible for the production of short-chain fatty acids (SCFA). It may be assumed, that SCFA are one of the mechanisms involved in the response to oral exposure to MP-PET.
Collapse
Affiliation(s)
- Ismena Gałęcka
- Department of Epizootiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719, Olsztyn, Poland.
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719, Olsztyn, Poland.
| | - Andrzej Rychlik
- Department of Clinical Diagnostics, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 14, 10-719, Olsztyn, Poland
| | - Jarosław Całka
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719, Olsztyn, Poland
| |
Collapse
|
3
|
Davias A, Lyon-Caen S, Rolland M, Iszatt N, Thomsen C, Sabaredzovic A, Sakhi AK, Monot C, Rayah Y, Ilhan ZE, Philippat C, Eggesbø M, Lepage P, Slama R. Associations between pre- and post-natal exposure to phthalate and DINCH metabolites and gut microbiota in one-year old children. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125204. [PMID: 39490662 DOI: 10.1016/j.envpol.2024.125204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
The gut microbiota is a collection of symbiotic microorganisms in the gastrointestinal tract. Its sensitivity to chemicals with widespread exposure, such as phthalates, is little known. We aimed to investigate the impact of perinatal exposure to phthalates on the infant gut microbiota at 12 months of age. Within SEPAGES cohort (Suivi de l'Exposition à la Pollution Atmosphérique durant la Grossesse et Effet sur la Santé), we assessed 13 phthalate metabolites and 2 di(isononyl) cyclohexane-1,2-dicarboxylate (DINCH) metabolites in repeated urine samples collected in pregnant women and their offspring. We obtained stool samples from 356 children at 12 months of age and sequenced the V3-V4 region of the 16S rRNA gene, allowing gut bacterial profiling. We used single-chemical (linear regressions) and mixture (BKMR, Bayesian Kernel Machine Regression) models to examine associations of phthalates and DINCH metabolites, with gut microbiota indices of α-diversity (specific richness and Shannon diversity) and the relative abundances of the most abundant microbiota phyla and genera. After correction for multiple testing, di(2-ethylhexyl) phthalate (ΣDEHP), diethyl phthalate (DEP) and bis(2-propylheptyl) phthalate (DPHP) metabolites 12-month urinary concentrations were associated with higher Shannon α-diversity of the child gut microbiota in single-chemical models. The multiple-chemical model (BKMR) suggested higher α-diversity with exposure to the phthalate mixture at 12 months, driven by the same phthalates. There were no associations between phthalate and DINCH exposure biomarkers at other time points and α-diversity after correction for multiple testing. ΣDEHP metabolites concentration at 12 months was associated with higher Coprococcus genus. Finally, ΣDEHP exposure at 12 months tended to be associated with higher phylum Firmicutes, an association not maintained after correction for multiple testing. Infancy exposure to phthalate might disrupt children's gut microbiota. The observed associations were cross-sectional, so that reverse causality cannot be excluded.
Collapse
Affiliation(s)
- Aline Davias
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France.
| | - Sarah Lyon-Caen
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France
| | - Matthieu Rolland
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France
| | - Nina Iszatt
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213, Oslo, Norway
| | - Cathrine Thomsen
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213, Oslo, Norway
| | - Azemira Sabaredzovic
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213, Oslo, Norway
| | - Amrit Kaur Sakhi
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213, Oslo, Norway
| | - Celine Monot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Yamina Rayah
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Zehra Esra Ilhan
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Claire Philippat
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France
| | - Merete Eggesbø
- Department of Clinical and Molecular Medicine, NTNU, Trondheim, Norway; Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 1432 Ås, Norway
| | - Patricia Lepage
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Rémy Slama
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France; SMILE, Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005, Paris, France; PARSEC, Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005, Paris, France
| |
Collapse
|
4
|
Hayat K, Zheng R, Zeng L, Ye Z, Pan J. Impact of Full-Spectrum and Infrared Lighting on Growth, Oxidative Stress, and Cecal Microbiota in Broilers. Antioxidants (Basel) 2024; 13:1442. [PMID: 39765771 PMCID: PMC11672630 DOI: 10.3390/antiox13121442] [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: 10/10/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
Lighting is crucial for the development of broilers as it affects their growth performance, oxidative stress, and overall health. This study investigates the impact of full-spectrum light, infrared light, and LED white light exposure on the growth performance, oxidative stress markers, and cecal microbiota of medium-growth yellow-feathered broilers. A total of 216 medium-growth yellow-feathered chicks (Yuhuang No. 5), five days old, were randomly divided into three groups: 72 chicks in each group, with three replicates of 24 chicks. The birds were raised under different lighting conditions, including LED infrared light (II), full-spectrum therapy light (FB), and LED white light (CG) until day 87. This experiment comprised the early growth phase and measured critical hormones such as Melatonin (Mel), Growth Hormone (GH), and Growth Hormone Releasing Hormone (GHRH), as well as Malondialdehyde (MDA), Superoxide Dismutase (SOD), and Catalase (CAT). Additionally, this study examined the differences in microbiota diversity and composition. The results demonstrated that LED infrared and full-spectrum light exposure significantly (p < 0.05) increased broiler body weight. Particularly, full-spectrum light was effective in comb redness and reducing final comb length and oxidative stress. Furthermore, full-spectrum light improved microbial prosperity and diversity compared with the other lighting conditions. Overall, the findings suggest that full-spectrum lighting is more beneficial for broiler growth, reducing oxidative stress, and promoting gut health compared with LED infrared lighting. These insights can be applied to optimizing broiler farming practices, thereby improving productivity and animal welfare.
Collapse
Affiliation(s)
- Khawar Hayat
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (K.H.)
- Ministry of Agriculture and Rural Affairs, Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | - Rongjin Zheng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (K.H.)
- Ministry of Agriculture and Rural Affairs, Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | - Li Zeng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (K.H.)
- Ministry of Agriculture and Rural Affairs, Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | - Zunzhong Ye
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (K.H.)
- Ministry of Agriculture and Rural Affairs, Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | - Jinming Pan
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (K.H.)
- Ministry of Agriculture and Rural Affairs, Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| |
Collapse
|
5
|
Taufer CR, da Silva J, Rampelotto PH. In Silico Analysis of Probiotic Bacteria Changes Across COVID-19 Severity Stages. Microorganisms 2024; 12:2353. [PMID: 39597740 PMCID: PMC11596909 DOI: 10.3390/microorganisms12112353] [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: 10/16/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
The gut microbiota plays a crucial role in modulating the immune response during COVID-19, with several studies reporting significant alterations in specific bacterial genera, including Akkermansia, Bacteroides, Bifidobacterium, Faecalibacterium, Lactobacillus, Oscillospira, and Ruminococcus. These genera are symbionts of the gut microbiota and contribute to host health. However, comparing results across studies is challenging due to differences in analysis methods and reference databases. We screened 16S rRNA raw datasets available in public databases on COVID-19, focusing on the V3-V4 region of the bacterial genome. In total, seven studies were included. All samples underwent the same bioinformatics pipeline, evaluating the differential abundance of these seven bacterial genera at each level of severity. The reanalysis identified significant changes in differential abundance. Bifidobacterium emerged as a potential biomarker of disease severity and a therapeutic target. Bacteroides presented a complex pattern, possibly related to disease-associated inflammation or opportunistic pathogen growth. Lactobacillus showed significant changes in abundance across the COVID-19 stages. On the other hand, Akkermansia and Faecalibacterium did not show significant differences, while Oscillospira and Ruminococcus produced statistically significant results but with limited relevance to COVID-19 severity. Our findings reveal new insights into the differential abundance of key bacterial genera in COVID-19, particularly Bifidobacterium and Bacteroides.
Collapse
Affiliation(s)
- Clarissa Reginato Taufer
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil; (C.R.T.); (J.d.S.)
| | - Juliana da Silva
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil; (C.R.T.); (J.d.S.)
- Graduate Program in Health and Human Development, Universidade La Salle, Canoas 92010-000, Brazil
| | - Pabulo Henrique Rampelotto
- Bioinformatics and Biostatistics Core Facility, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| |
Collapse
|
6
|
Ng CYJ, Zhong L, Ng HS, Goh KS, Zhao Y. Managing Type 2 Diabetes Mellitus via the Regulation of Gut Microbiota: A Chinese Medicine Perspective. Nutrients 2024; 16:3935. [PMID: 39599721 PMCID: PMC11597546 DOI: 10.3390/nu16223935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Type 2 Diabetes Mellitus (T2DM) is a metabolic disorder characterized by insulin resistance and inadequate insulin production. Given the increased frequency of T2DM and the health issues it can cause, there is an increasing need to develop alternative T2DM management strategies. One such approach is Chinese Medicine (CM), a complementary therapy widely used in T2DM treatment. Given the emphasis on gut microbiota in current research, studying CM in the treatment of T2DM via gut microbiota modulation could be beneficial. Scope and approach: The use of various CM methods for managing T2DM via gut microbiota modulation is highlighted in this review. Following an introduction of the gut microbiota and its role in T2DM pathogenesis, we will review the potential interactions between gut microbiota and T2DM. Thereafter, we will review various CM treatment modalities that modulate gut microbiota and provide perspectives for future research. Key findings and discussion: In T2DM, Akkermansia, Bifidobacterium, and Firmicutes are examples of gut microbiota commonly imbalanced. Studies have shown that CM therapies can modulate gut microbiota, leading to beneficial effects such as reduced inflammation, improved metabolism, and improved immunity. Among these treatment modalities, Chinese Herbal Medicine and acupuncture are the most well-studied, and several in vivo studies have demonstrated their potential in managing T2DM by modulating gut microbiota. However, the underlying biomolecular mechanisms of actions are not well elucidated, which is a key area for future research. Future studies could also investigate alternate CM therapies such as moxibustion and CM exercises and conduct large-scale clinical trials to validate their effectiveness in treatment.
Collapse
Affiliation(s)
- Chester Yan Jie Ng
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Linda Zhong
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Han Seong Ng
- Singapore General Hospital, Outram Rd., Singapore 169608, Singapore
- Academy of Chinese Medicine Singapore, 705 Serangoon Road, Singapore 328127, Singapore
| | - Kia Seng Goh
- Academy of Chinese Medicine Singapore, 705 Serangoon Road, Singapore 328127, Singapore
- Singapore College of Traditional Chinese Medicine, 640 Lor 4 Toa Payoh, Singapore 319522, Singapore
| | - Yan Zhao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Academy of Chinese Medicine Singapore, 705 Serangoon Road, Singapore 328127, Singapore
| |
Collapse
|
7
|
Dash D, Mishra V, Panda MK, Pathak SK. Effects of Lactobacillus spp. on Helicobacter pylori: A Promising Frontier in the Era of Antibiotic Resistance. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10396-z. [PMID: 39499454 DOI: 10.1007/s12602-024-10396-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 11/07/2024]
Abstract
Helicobacter pylori, a pathogenic bacterium responsible for multiple gastrointestinal disorders, has emerged as a major global concern due to rise in antibiotic resistance. Unwanted side effects of antibiotics therapy are further complicating the treatment strategies. Consequently, an alternative approach, using probiotics has emerged as a promising solution for treating H. pylori infections. Probiotics have shown considerable potential in increasing the cure rate and reducing the side effects through diverse mechanisms. Among the widely employed probiotics, Lactobacillus spp. has garnered particular attention in this review. After reviewing the studies on effects of Lactobacillus spp. on H. pylori, it is evident that several Lactobacillus spp. have demonstrated their potential efficacy against H. pylori infection, when administered alone or in conjunction with antibiotics, in a strain-specific manner. Furthermore, the inclusion of Lactobacillus spp. in the treatment regimen has also been associated with a reduction in the side effects related to antibiotic-based therapies. Future research may focus on identifying optimal strains and treatment regimens, understanding the long-term impacts of use, and determining their role in preventing H. pylori infection in various populations.
Collapse
Affiliation(s)
- Debabrata Dash
- Department of Biotechnology, Berhampur University, Bhanja Bihar, Berhampur, Odisha, 760007, India
| | - Vivek Mishra
- Department of Biotechnology, Berhampur University, Bhanja Bihar, Berhampur, Odisha, 760007, India
| | - Manoj Kumar Panda
- Department of Biotechnology, Berhampur University, Bhanja Bihar, Berhampur, Odisha, 760007, India
- Centre of Excellence on Bioprospecting of Ethno-Pharmaceuticals of Southern Odisha (CoE-BESO), Berhampur University, Bhanja Bihar, Berhampur, Odisha, 760007, India
| | - Sushil Kumar Pathak
- Department of Biotechnology, Berhampur University, Bhanja Bihar, Berhampur, Odisha, 760007, India.
- Centre of Excellence on Bioprospecting of Ethno-Pharmaceuticals of Southern Odisha (CoE-BESO), Berhampur University, Bhanja Bihar, Berhampur, Odisha, 760007, India.
| |
Collapse
|
8
|
Han Z, Li Y, Nan X, Zhou T, Li L, Li Y. Effect of probiotic supplementation combined with bismuth-containing quadruple therapy on gut microbiota during Helicobacter pylori eradication: a randomized, double-blind, placebo-controlled trial. Front Nutr 2024; 11:1484646. [PMID: 39479190 PMCID: PMC11521887 DOI: 10.3389/fnut.2024.1484646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024] Open
Abstract
Background Helicobacter pylori (H. pylori) eradication has been reported to affect gut microbiota distribution. This study aimed to evaluate the effect of probiotic supplementation on the gastrointestinal microbiota during eradication and the efficacy of bismuth-containing quadruple therapy. Methods One hundred treatment-naïve H. pylori-positive patients were randomly assigned 1:1 to receive 14-day bismuth-containing quadruple therapy (esomeprazole, bismuth, amoxicillin, and clarithromycin) combined with the probiotic (Bifidobacterium animalism subsp. lactis BLa80) or placebo. The Gastrointestinal Symptom Rating Scale (GSRS) was completed before and after treatment. Stool samples were collected for 16S rRNA gene sequencing at weeks 0, 2, and 10. Results No significant difference in the eradication rate was observed between the two groups. The incidence of adverse events, especially nausea (p = 0.029), was lower in the probiotic group. After treatment, the GSRS score decreased significantly in the probiotic group (p = 0.039). The gut microbiota underwent considerable changes immediately following eradication treatment, predominantly characterized by an increase in Proteobacteria at the expense of commensal Firmicutes and Bacteroidota, but gradually returned to baseline after eight weeks. By week 10, beneficial genera such as Lachnoclostridium, Parasutterella, Hungatella, and Akkermansia were notably enriched in the probiotic group. Additionally, the correlation networks in the probiotic group were closer to their initial levels at week 10 compared to the placebo group. Conclusion Disturbances in the gut microbiota following H. pylori treatment appeared to be temporary, and probiotic supplementation could mitigate antibiotic-induced alterations in the gut microbiota. This study also provided evidence supporting the effectiveness of probiotics in alleviating gastrointestinal symptoms.
Collapse
Affiliation(s)
- Zhongxue Han
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, China
| | - Yueyue Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, China
| | - Xueping Nan
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, China
| | - Tao Zhou
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, China
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Lixiang Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, China
| | - Yanqing Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Jinan, China
| |
Collapse
|
9
|
Horwell E, Bearn P, Cutting SM. A microbial symphony: a literature review of the factors that orchestrate the colonization dynamics of the human colonic microbiome during infancy and implications for future health. MICROBIOME RESEARCH REPORTS 2024; 4:1. [PMID: 40207275 PMCID: PMC11977369 DOI: 10.20517/mrr.2024.32] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Since the advent of new sequencing and bioinformatic technologies, our understanding of the human microbiome has expanded rapidly over recent years. Numerous studies have indicated causal links between alterations to the microbiome and a range of pathological conditions. Furthermore, a large body of epidemiological data is starting to suggest that exposure, or lack thereof, to specific microbial species during the first five years of life has key implications for long-term health outcomes. These include chronic inflammatory and metabolic conditions such as diabetes, asthma, inflammatory bowel disease (IBD), and obesity, with the effects lasting into adulthood. Human microbial colonisation during these first five years of life is a highly dynamic process, with multiple environmental exposures recently being characterised to have influence before the microbiome stabilises and resembles that of an adult at 3-5 years. This short period of time, known as the window of opportunity, appears to "prime" immunoregulation for later life. Understanding and appreciating this aspect of human physiology is therefore crucial for clinicians, scientists, and public health officials. This review outlines the most recent evidence for the pre- and post-natal environments that order the development of the microbiome, how these influences metabolic and immunoregulatory pathways, and their associated health outcomes. It also discusses the limitations of the current knowledge base, and describes the potential microbiome-mediated interventions and public health measures that may have therapeutic potential in the future.
Collapse
Affiliation(s)
- Edward Horwell
- Department of Biomedical Sciences, The Bourne Laboratory, Royal Holloway University of London, London TW20 0EX, UK
- Department of Colorectal Surgery, Ashford and Saint Peter’s NHS Foundation Trust, London KT16 0PZ, UK
| | - Philip Bearn
- Department of Colorectal Surgery, Ashford and Saint Peter’s NHS Foundation Trust, London KT16 0PZ, UK
| | - Simon M. Cutting
- Department of Biomedical Sciences, The Bourne Laboratory, Royal Holloway University of London, London TW20 0EX, UK
| |
Collapse
|
10
|
Davias A, Lyon-Caen S, Rolland M, Iszatt N, Thomsen C, Haug LS, Sakhi AK, Monot C, Rayah Y, Ilhan ZE, Jovanovic N, Philippat C, Eggesbo M, Lepage P, Slama R. Perinatal Exposure to Phenols and Poly- and Perfluoroalkyl Substances and Gut Microbiota in One-Year-Old Children. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:15395-15414. [PMID: 39173114 DOI: 10.1021/acs.est.3c09927] [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: 08/24/2024]
Abstract
The role of the gut microbiota in human health calls for a better understanding of its determinants. In particular, the possible effects of chemicals with widespread exposure other than pharmaceuticals are little known. Our aim was to characterize the sensitivity of the early-life gut microbiota to specific chemicals with possible antimicrobial action. Within the SEPAGES French couple-child cohort, we assessed 12 phenols in repeated urine samples from 356 pregnant women and their offspring and 19 poly- and perfluoroalkyl substances (PFASs) in serum from the pregnant women. We collected stool samples from the children at one year of age, in which the V3-V4 region of the 16S rRNA gene was sequenced, allowing for gut bacterial profiling. Associations of each chemical with α- and β-diversity indices of the gut microbiota and with the relative abundance of the most abundant taxa were assessed using single-pollutant and mixture (BKMR) models. Perinatal exposure to certain parabens was associated with gut microbiota α- and β-diversity and with Firmicutes and Proteobacteria. Suggestive associations of certain phenols with genera of the Lachnospiraceae and Enterobacteriaceae families were observed, but these were not maintained after correction for multiple testing. Parabens, which have known antimicrobial properties, might disrupt the child gut microbiota, but larger studies are required to confirm these findings.
Collapse
Affiliation(s)
- Aline Davias
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Sarah Lyon-Caen
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Matthieu Rolland
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Nina Iszatt
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Cathrine Thomsen
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Line Småstuen Haug
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Amrit Kaur Sakhi
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Celine Monot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Yamina Rayah
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Zehra Esra Ilhan
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Nicolas Jovanovic
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Claire Philippat
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Merete Eggesbo
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Patricia Lepage
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Rémy Slama
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| |
Collapse
|
11
|
Hick E, Suárez M, Rey A, Mantecón L, Fernández N, Solís G, Gueimonde M, Arboleya S. Personalized Nutrition with Banked Human Milk for Early Gut Microbiota Development: In Pursuit of the Perfect Match. Nutrients 2024; 16:1976. [PMID: 38999725 PMCID: PMC11243202 DOI: 10.3390/nu16131976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
The correct initial colonization and establishment of the gut microbiota during the early stages of life is a key step, with long-lasting consequences throughout the entire lifespan of the individual. This process is affected by several perinatal factors; among them, feeding mode is known to have a critical role. Breastfeeding is the optimal nutrition for neonates; however, it is not always possible, especially in cases of prematurity or early pathology. In such cases, most commonly babies are fed with infant formulas in spite of the official nutritional and health international organizations' recommendation on the use of donated human milk through milk banks for these cases. However, donated human milk still does not totally match maternal milk in terms of infant growth and gut microbiota development. The present review summarizes the practices of milk banks and hospitals regarding donated human milk, its safety and quality, and the health outcomes in infants fed with donated human milk. Additionally, we explore different alternatives to customize pasteurized donated human milk with the aim of finding the perfect match between each baby and banked milk for promoting the establishment of a beneficial gut microbiota from the early stages of life.
Collapse
Affiliation(s)
- Emilia Hick
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
| | - Marta Suárez
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Alejandra Rey
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
| | - Laura Mantecón
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Nuria Fernández
- Pediatrics Service, University Hospital of Cabueñes (CAB-SESPA), 33394 Gijón, Spain
| | - Gonzalo Solís
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
12
|
Mohite SV, Sharma KK. Gut microbial metalloproteins and its role in xenobiotics degradation and ROS scavenging. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:495-538. [PMID: 38960484 DOI: 10.1016/bs.apcsb.2024.03.004] [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: 07/05/2024]
Abstract
The gut microbial metalloenzymes play an important role in maintaining the balance between gut microbial ecosystem, human physiologically processes and immune system. The metals coordinated into active site contribute in various detoxification and defense strategies to avoid unfavourable environment and ensure bacterial survival in human gut. Metallo-β-lactamase is a potent degrader of antibiotics present in periplasmic space of both commensals and pathogenic bacteria. The resistance to anti-microbial agents developed in this enzyme is one of the global threats for human health. The organophosphorus eliminator, organophosphorus hydrolases have evolved over a course of time to hydrolyze toxic organophosphorus compounds and decrease its effect on human health. Further, the redox stress responders namely superoxide dismutase and catalase are key metalloenzymes in reducing both endogenous and exogenous oxidative stress. They hold a great importance for pathogens as they contribute in pathogenesis in human gut along with reduction of oxidative stress. The in-silico study on these enzymes reveals the importance of point mutation for the evolution of these enzymes in order to enhance their enzyme activity and stability. Various mutation studies were conducted to investigate the catalytic activity of these enzymes. By using the "directed evolution" method, the enzymes involved in detoxification and defense system can be engineered to produce new variants with enhance catalytic features, which may be used to predict the severity due to multi-drug resistance and degradation pattern of organophosphorus compounds in human gut.
Collapse
Affiliation(s)
- Shreya Vishwas Mohite
- Laboratory of Enzymology and Gut Microbiology, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Krishna Kant Sharma
- Laboratory of Enzymology and Gut Microbiology, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, India.
| |
Collapse
|
13
|
Gajda-Morszewski P, Poznańska A, Federyga E, Ściuk A, Brindell M. Encapsulated Mn-Saturated Lactoferrin as a Safe Source of Manganese Ions for Restoring Probiotic Lactobacillus plantarum. Molecules 2024; 29:2735. [PMID: 38930801 PMCID: PMC11205955 DOI: 10.3390/molecules29122735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
The growth of Lactobacillus plantarum, a member of the Lactobacillus genus, which plays a crucial role in the bacterial microbiome of the gut, is significantly influenced by manganese ions. They can be safely delivered to the intestines by exploiting the chelating abilities of lactoferrin. The aim of this work was to encapsulate lactoferrin saturated with manganese ions (MnLf) in a system based on the Eudragit® RS polymer to protect protein from degradation and manganese release in the gastric environment. The entrapment efficiency was satisfactory, reaching about 95%, and most importantly, manganese ions were not released during microparticles (MPs) formation. The release profile of the protein from the freshly prepared MPs was sustained, with less than 15% of the protein released within the first hour. To achieve similar protein release efficiency, freeze-drying was carried out in the presence of 10% (w/v) mannitol as a cryoprotectant for MPs frozen at -20 °C. MPs with encapsulated MnLf exhibited prebiotic activity towards Lactobacillus plantarum. More importantly, the presence of equivalent levels of manganese ions in free form in the medium, as well as chelating by lactoferrin encapsulated in MPs, had a similar impact on stimulating bacterial growth. This indicates that the bioavailability of manganese ions in our prepared system is very good.
Collapse
Affiliation(s)
- Przemysław Gajda-Morszewski
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University in Krakow, Gronostajowa 2, 30-387 Kraków, Poland; (P.G.-M.); (A.P.); (E.F.)
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Prof. St. Łojasiewicza St 11, 30-348 Kraków, Poland;
| | - Anna Poznańska
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University in Krakow, Gronostajowa 2, 30-387 Kraków, Poland; (P.G.-M.); (A.P.); (E.F.)
| | - Eryk Federyga
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University in Krakow, Gronostajowa 2, 30-387 Kraków, Poland; (P.G.-M.); (A.P.); (E.F.)
| | - Anna Ściuk
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Prof. St. Łojasiewicza St 11, 30-348 Kraków, Poland;
- Department of Crystal Chemistry and Crystal Physics, Faculty of Chemistry, Jagiellonian University in Krakow, Gronostajowa 2, 30-387 Kraków, Poland
| | - Małgorzata Brindell
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University in Krakow, Gronostajowa 2, 30-387 Kraków, Poland; (P.G.-M.); (A.P.); (E.F.)
| |
Collapse
|
14
|
Jurek JM, Castro-Marrero J. A Narrative Review on Gut Microbiome Disturbances and Microbial Preparations in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Implications for Long COVID. Nutrients 2024; 16:1545. [PMID: 38892479 PMCID: PMC11173566 DOI: 10.3390/nu16111545] [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: 04/29/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Myalgic encephalomyelitis, also known as chronic fatigue syndrome (ME/CFS), and long COVID are complex, multisystemic and long-term disabling conditions characterized by debilitating post-exertional malaise and other core symptoms related to immune dysregulation resultant from post-viral infection, including mitochondrial dysfunction, chronic neuroinflammation and gut dysbiosis. The reported associations between altered microbiota composition and cardinal symptoms of ME/CFS and long COVID suggest that the use of microbial preparations, such as probiotics, by restoring the homeostasis of the brain-immune-gut axis, may help in the management of symptoms in both conditions. Therefore, this review aims to investigate the implications of alerted gut microbiome and assess the evidence supporting use of microbial-based preparations, including probiotics, synbiotics, postbiotics alone and/or in combination with other nutraceuticals in the management of fatigue, inflammation and neuropsychiatric and gastrointestinal symptoms among patients with ME/CFS and long COVID.
Collapse
Affiliation(s)
- Joanna Michalina Jurek
- Unit of Research in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Long COVID, Rheumatology Research Division, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
- Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43005 Tarragona, Spain
| | - Jesus Castro-Marrero
- Unit of Research in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Long COVID, Rheumatology Research Division, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| |
Collapse
|
15
|
Liu B, Jiang L, Liu Y, Sun H, Yan J, Kang C, Yang B. Enterohaemorrhagic E. coli utilizes host- and microbiota-derived L-malate as a signaling molecule for intestinal colonization. Nat Commun 2023; 14:7227. [PMID: 37945607 PMCID: PMC10636207 DOI: 10.1038/s41467-023-43149-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
The mammalian gastrointestinal tract is a complex environment that hosts a diverse microbial community. To establish infection, bacterial pathogens must be able to compete with the indigenous microbiota for nutrients, as well as sense the host environment and modulate the expression of genes essential for colonization and virulence. Here, we found that enterohemorrhagic Escherichia coli (EHEC) O157:H7 imports host- and microbiota-derived L-malate using the DcuABC transporters and converts these substrates into fumarate to fuel anaerobic fumarate respiration during infection, thereby promoting its colonization of the host intestine. Moreover, L-malate is important not only for nutrient metabolism but also as a signaling molecule that activates virulence gene expression in EHEC O157:H7. The complete virulence-regulating pathway was elucidated; the DcuS/DcuR two-component system senses high L-malate levels and transduces the signal to the master virulence regulator Ler, which in turn activates locus of enterocyte effacement (LEE) genes to promote EHEC O157:H7 adherence to epithelial cells of the large intestine. Disruption of this virulence-regulating pathway by deleting either dcuS or dcuR significantly reduced colonization by EHEC O157:H7 in the infant rabbit intestinal tract; therefore, targeting these genes and altering physiological aspects of the intestinal environment may offer alternatives for EHEC infection treatment.
Collapse
Affiliation(s)
- Bin Liu
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, 300457, P. R. China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, 300071, P. R. China
| | - Lingyan Jiang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, 300457, P. R. China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, 300071, P. R. China
| | - Yutao Liu
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, 300457, P. R. China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, 300071, P. R. China
| | - Hongmin Sun
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, 300457, P. R. China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, 300071, P. R. China
| | - Jun Yan
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, 300457, P. R. China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, 300071, P. R. China
| | - Chenbo Kang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, 300457, P. R. China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, 300071, P. R. China
| | - Bin Yang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, 300457, P. R. China.
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, 300071, P. R. China.
| |
Collapse
|
16
|
Gomes N, Ferreira-Sa L, Alves N, Dallago B, Moraes A, Carvalho JL, Nitz N, Hagström L, Braz S, Machado ER, Gurgel-Gonçalves R, Hecht M. Uncovering the effects of Giardia duodenalis on the balance of DNA viruses and bacteria in children's gut microbiota. Acta Trop 2023; 247:107018. [PMID: 37673134 DOI: 10.1016/j.actatropica.2023.107018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/08/2023]
Abstract
The neglected parasitosis giardiasis is one of the most common intestinal infections worldwide, affecting mainly infants and young children. Giardia duodenalis may disturb the local microbiome, leading to intestinal ecosystem disorders, and altering different processes in the host, such as the immune response. Nevertheless, the alterations promoted by G. duodenalis on the human gut microbiome have not been thoroughly investigated. Here, we characterized the gut microbiota of G. duodenalis-infected children and determine the main alterations promoted by the parasite. To do so, fecal samples of 26 infected and four uninfected children aged 2 to 6 years old were processed for High Efficiency Microarray analysis, in order to describe their bacterial and viral profiles. Then, we quantified the total bacterial population by qPCR and assessed fecal calprotectin levels, which are closely related with gut inflammation. A total of 286 bacteria's species and 17 viruses' strains were identified. Our results revealed no statistically significant differences between G. duodenalis positive and negative groups in the taxa's phyla and families. However, bacterial species diversity was increased in children infected with G. duodenalis (p < 0.05), while the total number of bacteria was decreased (p < 0.05). Considering the virome analysis, 17 different strains were identified, 88% being bacteriophages. The correlation analysis revealed an important disruption in the balance of DNA virus and bacteria within the intestinal microbiota of Giardia-positive children. Our findings constitute the first description of the gut virome of Giardia-infected children and suggest that G. duodenalis infection exerts a modulatory effect on the gut microbiome, promoting local inflammation and altering the equilibrium of the parasite-microbiota-host triad. This highlights the importance of considering polymicrobial associations and understanding the broader context of giardiasis. Overall, our study provides new insights into the complex interactions between intestinal parasites and the microbiota, which may have implications for the development of novel therapeutic interventions in the future.
Collapse
Affiliation(s)
- Nélio Gomes
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil
| | - Lana Ferreira-Sa
- Laboratory of Medical Parasitology and Vector Biology Faculty of Medicine, University of Brasília, Brasília, Brazil
| | - Nayra Alves
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil
| | - Bruno Dallago
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil
| | - Aline Moraes
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil
| | - Juliana Lott Carvalho
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil; Genomic Sciences and Biotechnology Program, Catholic University of Brasília, Distrito Federal, Brazil
| | - Nadjar Nitz
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil
| | - Luciana Hagström
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil
| | - Shélida Braz
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil
| | - Eleuza Rodrigues Machado
- Laboratory of Medical Parasitology and Vector Biology Faculty of Medicine, University of Brasília, Brasília, Brazil
| | - Rodrigo Gurgel-Gonçalves
- Laboratory of Medical Parasitology and Vector Biology Faculty of Medicine, University of Brasília, Brasília, Brazil
| | - Mariana Hecht
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil.
| |
Collapse
|
17
|
Okoshi K, Sakurai K, Yamamoto M, Mori C. Maternal antibiotic exposure and childhood allergies: The Japan Environment and Children's Study. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100137. [PMID: 37781654 PMCID: PMC10509907 DOI: 10.1016/j.jacig.2023.100137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 10/03/2023]
Abstract
Background The association of maternal antibiotic exposure during pregnancy with childhood allergic diseases remains unclear. Objective We aimed to evaluate the association of maternal exposure to antibiotic use during pregnancy with childhood allergic diseases up to the age of 3 years by using data from a large Japanese birth cohort. Methods We analyzed data on 78,678 pregnant women and their offspring aged 0 to 3 years. Prenatal antibiotic exposure was defined as the use of any antimicrobial agent during pregnancy. Information was collected from maternal interviews and medical record transcripts. The outcome variables in this study included preschool asthma, wheezing, food allergy, atopic dermatitis, eczema, allergic rhinoconjunctivitis, and any allergic disease. We used logistic regression analysis to evaluate the association of antibiotic exposure during pregnancy with childhood allergic diseases. Results Among the participating mothers, 28.5% used antibiotics during pregnancy. Antibiotic exposure during pregnancy was associated with preschool asthma (adjusted odds ratio [aOR] = 1.12 [95% CI = 1.06-1.19]), wheezing (aOR = 1.11 [95% CI = 1.07-1.15]), allergic rhinoconjunctivitis (aOR = 1.10 [95% CI = 1.03-1.17]) and any allergic disease (aOR = 1.09 [95% CI = 1.05-1.14]) in offspring up to age 3 years. In contrast, maternal antibiotic use was not associated with food allergies, atopic dermatitis, or eczema. Additionally, the significant associations were not influenced by the timing of antibiotic exposure, sex of the infants, or maternal history of allergies. Conclusion Maternal antibiotic exposure during pregnancy is associated with an increased risk of childhood respiratory allergies.
Collapse
Affiliation(s)
- Kouta Okoshi
- Department of Sustainable Health Science, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Innovation Center, Central Research Laboratory, NIPPN Corporation, Kanagawa, Japan
| | - Kenichi Sakurai
- Department of Nutrition and Metabolic Medicine, Center for Preventive Medical Sciences, Chiba University, Japan
| | - Midori Yamamoto
- Department of Sustainable Health Science, Center for Preventive Medical Sciences, Chiba University, Japan
| | - Chisato Mori
- Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Sustainable Health Science, Center for Preventive Medical Sciences, Chiba University, Japan
| |
Collapse
|
18
|
Kundu S, Nayak S, Rakshit D, Singh T, Shukla R, Khatri DK, Mishra A. The microbiome-gut-brain axis in epilepsy: pharmacotherapeutic target from bench evidence for potential bedside applications. Eur J Neurol 2023; 30:3557-3567. [PMID: 36880679 DOI: 10.1111/ene.15767] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
The gut-brain axis augments the bidirectional communication between the gut and brain and modulates gut homeostasis and the central nervous system through the hypothalamic-pituitary-adrenal axis, enteroendocrine system, neuroendocrine system, inflammatory and immune pathways. Preclinical and clinical reports showed that gut dysbiosis might play a major regulatory role in neurological diseases such as epilepsy, Parkinson's, multiple sclerosis, and Alzheimer's disease. Epilepsy is a chronic neurological disease that causes recurrent and unprovoked seizures, and numerous risk factors are implicated in developing epilepsy. Advanced consideration of the gut-microbiota-brain axis can reduce ambiguity about epilepsy pathology, antiepileptic drugs, and effective therapeutic targets. Gut microbiota sequencing analysis reported that the level of Proteobacteria, Verrucomicrobia, Fusobacteria, and Firmicutes was increased and the level of Actinobacteria and Bacteroidetes was decreased in epilepsy patients. Clinical and preclinical studies also indicated that probiotics, ketogenic diet, faecal microbiota transplantation, and antibiotics can improve gut dysbiosis and alleviate seizure by enhancing the abundance of healthy biota. This study aims to give an overview of the connection between gut microbiota, and epilepsy, how gut microbiome changes may cause epilepsy, and whether gut microbiome restoration could be used as a treatment for epilepsy.
Collapse
Affiliation(s)
- Snehashis Kundu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Sudipta Nayak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Debarati Rakshit
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Awanish Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| |
Collapse
|
19
|
Sarker P, Mitro A, Hoque H, Hasan MN, Nurnabi Azad Jewel GM. Identification of potential novel therapeutic drug target against Elizabethkingia anophelis by integrative pan and subtractive genomic analysis: An in silico approach. Comput Biol Med 2023; 165:107436. [PMID: 37690289 DOI: 10.1016/j.compbiomed.2023.107436] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 07/08/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023]
Abstract
Elizabethkingia anophelis is a human pathogen responsible for severe nosocomial infections in neonates and immunocompromised patients. The significantly higher mortality rate from E. anophelis infections and the lack of available regimens highlight the critical need to explore novel drug targets. The current study investigated effective novel drug targets by employing a comprehensive in silico subtractive genomic approach integrated with pangenomic analysis of E. anophelis strains. A total of 2809 core genomic proteins were found by pangenomic analysis of non-paralogous proteins. Subsequently, 156 pathogen-specific, 442 choke point, 202 virulence factor, 53 antibiotic resistant and 119 host-pathogen interacting proteins were identified in E. anophelis. By subtractive genomic approach, at first 791 proteins were found to be indispensable for the survival of E. anophelis. 558 and 315 proteins were detected as non-homologous to human and gut microflora respectively. Following that 245 cytoplasmic, 245 novel, and 23 broad-spectrum targets were selected and finally four proteins were considered as potential therapeutic targets of E. anophelis based on highest degree score in PPI network. Among those, three proteins were subjected to molecular docking and subsequent MD simulation as one protein did not contain a plausible binding pocket with sufficient surface area and volume. All the complexes were found to be stable and compact in 100 ns molecular dynamics simulation studies as measured by RMSD, RMSF, and Rg. These three short-listed targets identified in this study may lead to the development of novel antimicrobials capable of curing infections and pave the way to prevent and control the disease progression caused by the deadly agent E. anophelis.
Collapse
Affiliation(s)
- Parth Sarker
- Dept. of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, University Ave, Sylhet-3114, Bangladesh; Computational Biology and Bioinformatics Lab, Dept. of GEB, SUST, Sylhet-3114, Bangladesh
| | - Arnob Mitro
- Dept. of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, University Ave, Sylhet-3114, Bangladesh; Computational Biology and Bioinformatics Lab, Dept. of GEB, SUST, Sylhet-3114, Bangladesh
| | - Hammadul Hoque
- Dept. of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, University Ave, Sylhet-3114, Bangladesh
| | - Md Nazmul Hasan
- Dept. of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, University Ave, Sylhet-3114, Bangladesh
| | - G M Nurnabi Azad Jewel
- Dept. of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, University Ave, Sylhet-3114, Bangladesh; Computational Biology and Bioinformatics Lab, Dept. of GEB, SUST, Sylhet-3114, Bangladesh.
| |
Collapse
|
20
|
De Marco G, Cappello T, Maisano M. Histomorphological Changes in Fish Gut in Response to Prebiotics and Probiotics Treatment to Improve Their Health Status: A Review. Animals (Basel) 2023; 13:2860. [PMID: 37760260 PMCID: PMC10525268 DOI: 10.3390/ani13182860] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/06/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
The gastrointestinal tract (GIT) promotes the digestion and absorption of feeds, in addition to the excretion of waste products of digestion. In fish, the GIT is divided into four regions, the headgut, foregut, midgut, and hindgut, to which glands and lymphoid tissues are associated to release digestive enzymes and molecules involved in the immune response and control of host-pathogens. The GIT is inhabited by different species of resident microorganisms, the microbiota, which have co-evolved with the host in a symbiotic relationship and are responsible for metabolic benefits and counteracting pathogen infection. There is a strict connection between a fish's gut microbiota and its health status. This review focuses on the modulation of fish microbiota by feed additives based on prebiotics and probiotics as a feasible strategy to improve fish health status and gut efficiency, mitigate emerging diseases, and maximize rearing and growth performance. Furthermore, the use of histological assays as a valid tool for fish welfare assessment is also discussed, and insights on nutrient absorptive capacity and responsiveness to pathogens in fish by gut morphological endpoints are provided. Overall, the literature reviewed emphasizes the complex interactions between microorganisms and host fish, shedding light on the beneficial use of prebiotics and probiotics in the aquaculture sector, with the potential to provide directions for future research.
Collapse
Affiliation(s)
| | - Tiziana Cappello
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (G.D.M.); (M.M.)
| | | |
Collapse
|
21
|
Fraboul J, Biroli G, De Monte S. Artificial selection of communities drives the emergence of structured interactions. J Theor Biol 2023; 571:111557. [PMID: 37302465 DOI: 10.1016/j.jtbi.2023.111557] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/07/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023]
Abstract
Species-rich communities, such as the microbiota or microbial ecosystems, provide key functions for human health and climatic resilience. Increasing effort is being dedicated to design experimental protocols for selecting community-level functions of interest. These experiments typically involve selection acting on populations of communities, each of which is composed of multiple species. If numerical simulations started to explore the evolutionary dynamics of this complex, multi-scale system, a comprehensive theoretical understanding of the process of artificial selection of communities is still lacking. Here, we propose a general model for the evolutionary dynamics of communities composed of a large number of interacting species, described by disordered generalised Lotka-Volterra equations. Our analytical and numerical results reveal that selection for scalar community functions leads to the emergence, along an evolutionary trajectory, of a low-dimensional structure in an initially featureless interaction matrix. Such structure reflects the combination of the properties of the ancestral community and of the selective pressure. Our analysis determines how the speed of adaptation scales with the system parameters and the abundance distribution of the evolved communities. Artificial selection for larger total abundance is thus shown to drive increased levels of mutualism and interaction diversity. Inference of the interaction matrix is proposed as a method to assess the emergence of structured interactions from experimentally accessible measures.
Collapse
Affiliation(s)
- Jules Fraboul
- Laboratoire de Physique de l'École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université de Paris, Paris, F-75005, France.
| | - Giulio Biroli
- Laboratoire de Physique de l'École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université de Paris, Paris, F-75005, France
| | - Silvia De Monte
- Institut de Biologie de l'ENS (IBENS), Département de Biologie, Ecole normale supérieure, CNRS, INSERM, Université PSL, Paris, 75005, France; Department of Evolutionary Theory, Max Planck Institute for Evolutionary Biology, Plön, Germany
| |
Collapse
|
22
|
Ali H, Samad A, Ajmal A, Ali A, Ali I, Danial M, Kamal M, Ullah M, Ullah R, Kalim M. Identification of Drug Targets and Their Inhibitors in Yersinia pestis Strain 91001 through Subtractive Genomics, Machine Learning, and MD Simulation Approaches. Pharmaceuticals (Basel) 2023; 16:1124. [PMID: 37631039 PMCID: PMC10459760 DOI: 10.3390/ph16081124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/20/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Yersinia pestis, the causative agent of plague, is a Gram-negative bacterium. If the plague is not properly treated it can cause rapid death of the host. Bubonic, pneumonic, and septicemic are the three types of plague described. Bubonic plague can progress to septicemic plague, if not diagnosed and treated on time. The mortality rate of pneumonic and septicemic plague is quite high. The symptom-defining disease is the bubo, which is a painful lymph node swelling. Almost 50% of bubonic plague leads to sepsis and death if not treated immediately with antibiotics. The host immune response is slow as compared to other bacterial infections. Clinical isolates of Yersinia pestis revealed resistance to many antibiotics such as tetracycline, spectinomycin, kanamycin, streptomycin, minocycline, chloramphenicol, and sulfonamides. Drug discovery is a time-consuming process. It always takes ten to fifteen years to bring a single drug to the market. In this regard, in silico subtractive proteomics is an accurate, rapid, and cost-effective approach for the discovery of drug targets. An ideal drug target must be essential to the pathogen's survival and must be absent in the host. Machine learning approaches are more accurate as compared to traditional virtual screening. In this study, k-nearest neighbor (kNN) and support vector machine (SVM) were used to predict the active hits against the beta-ketoacyl-ACP synthase III drug target predicted by the subtractive genomics approach. Among the 1012 compounds of the South African Natural Products database, 11 hits were predicted as active. Further, the active hits were docked against the active site of beta-ketoacyl-ACP synthase III. Out of the total 11 active hits, the 3 lowest docking score hits that showed strong interaction with the drug target were shortlisted along with the standard drug and were simulated for 100 ns. The MD simulation revealed that all the shortlisted compounds display stable behavior and the compounds formed stable complexes with the drug target. These compounds may have the potential to inhibit the beta-ketoacyl-ACP synthase III drug target and can help to combat Yersinia pestis-related infections. The dataset and the source codes are freely available on GitHub.
Collapse
Affiliation(s)
- Hamid Ali
- Department of Biosciences, COMSATS University Islamabad, Park Road, Tarlai Kalan, Islamabad 44000, Pakistan
| | - Abdus Samad
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan; (A.S.); (A.A.); (M.D.); (M.K.)
| | - Amar Ajmal
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan; (A.S.); (A.A.); (M.D.); (M.K.)
| | - Amjad Ali
- Faculty of Biological Sciences, Department of Biochemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - Ijaz Ali
- Centre for Applied Mathematics and Bioinformatics (CAMB), Gulf University for Science and Technology, Hawally 32093, Kuwait;
| | - Muhammad Danial
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan; (A.S.); (A.A.); (M.D.); (M.K.)
| | - Masroor Kamal
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan; (A.S.); (A.A.); (M.D.); (M.K.)
| | - Midrar Ullah
- Department of Biotechnology, Shaheed Benazir Bhutto University Sheringal, Dir Upper 18050, Pakistan;
| | - Riaz Ullah
- Department of Pharmacognosy, College of Pharmacy King Saud University, Riyadh 11451, Saudi Arabia;
| | - Muhammad Kalim
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA;
- Houston Methodist Cancer Center/Weill Cornel Medicine, Houston, TX 77030, USA
| |
Collapse
|
23
|
Novoa Rama E, Bailey M, Kumar S, Leone C, den Bakker HC, Thippareddi H, Singh M. Characterizing the gut microbiome of broilers raised under conventional and no antibiotics ever practices. Poult Sci 2023; 102:102832. [PMID: 37343348 PMCID: PMC10404755 DOI: 10.1016/j.psj.2023.102832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/23/2023] Open
Abstract
Meat from broilers raised without the use of antibiotics is becoming increasingly popular among consumers. Consequently, interest in the microbial profiling of chickens produced under nonconventional practices is growing, however, research on this topic is lacking. The current study was designed to characterize the dynamics of gut microbial populations of broilers raised under conventional and no antibiotics ever (NAE) practices. Four commercial farms (2 conventional and 2 NAE) were included in this study. On each farm, cecal (n = 224) and ileal (n = 224) contents were collected from birds at different stages during the grow out of a single flock and following transportation to the processing facility. Cecal microbiota was dominated by the genera Escherichia and Enterococcus upon hatching in both conventional and NAE flocks, shifting with time toward predominantly Faecalibacterium and Bacteroides. The composition of cecal microbial communities of NAE broilers was different than that of conventional chickens (P ≤ 0.05). Conventional broilers harbored a rich, but less diverse cecal microbiota than NAE, while the ileal microbiota was primarily populated with genera previously named Lactobacillus, which exhibited a higher abundance in NAE broilers (P ≤ 0.05). In both production systems, the microbiota followed a similar temporal succession that was more evident in the ceca. Transportation to the processing plant impacted the microbial composition of the ileum (P ≤ 0.05), characterized by an increase in the relative abundance of Psychrobacter. Finally, differential abundance analysis showed a positive correlation between Campylobacter and Enorma within the cecum microbiota, and a negative correlation with Salmonella.
Collapse
Affiliation(s)
- Estefanía Novoa Rama
- Department of Food Science and Technology, University of Georgia, Athens, GA, USA
| | - Matthew Bailey
- Department of Poultry Science, Auburn University, Auburn, AL, USA
| | - Sanjay Kumar
- Department of Poultry Science, University of Georgia, Athens, GA, USA
| | - Cortney Leone
- Department of Poultry Science, University of Georgia, Athens, GA, USA
| | - Henk C den Bakker
- Department of Food Science and Technology, University of Georgia, Athens, GA, USA; Center for Food Safety, Department of Food Science and Technology, University of Georgia, Griffin, GA, USA.
| | | | - Manpreet Singh
- Department of Food Science and Technology, University of Georgia, Athens, GA, USA
| |
Collapse
|
24
|
Guo GJ, Yao F, Lu WP, Xu HM. Gut microbiome and metabolic-associated fatty liver disease: Current status and potential applications. World J Hepatol 2023; 15:867-882. [PMID: 37547030 PMCID: PMC10401411 DOI: 10.4254/wjh.v15.i7.867] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/11/2023] [Accepted: 06/30/2023] [Indexed: 07/21/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is one of the most common chronic liver diseases worldwide. In recent years, the occurrence rate of MAFLD has been on the rise, mainly due to lifestyle changes, high-calorie diets, and imbalanced dietary structures, thereby posing a threat to human health and creating heavy social and economic burdens. With the development of 16S sequencing and integrated multi-omics analysis, the role of the gut microbiota (GM) and its metabolites in MAFLD has been further recognized. The GM plays a role in digestion, energy metabolism, vitamin synthesis, the prevention of pathogenic bacteria colonisation, and immunoregulation. The gut-liver axis is one of the vital links between the GM and the liver. Toxic substances in the intestine can enter the liver through the portal vascular system when the intestinal barrier is severely damaged. The liver also influences the GM in various ways, such as bile acid circulation. The gut-liver axis is essential in maintaining the body's normal physiological state and plays a role in the onset and prognosis of many diseases, including MAFLD. This article reviews the status of the GM and MAFLD and summarizes the GM characteristics in MAFLD. The relationship between the GM and MAFLD is discussed in terms of bile acid circulation, energy metabolism, micronutrients, and signalling pathways. Current MAFLD treatments targeting the GM are also listed.
Collapse
Affiliation(s)
- Gong-Jing Guo
- Gastroenterology Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen 518172, Guangdong Province, China
| | - Fei Yao
- Department of Science and Education, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, Guangdong Province, China
| | - Wei-Peng Lu
- The First Clinical School, Guangzhou Medical University, Guangzhou 510120, Guangdong Province, China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, Guangdong Province, China.
| |
Collapse
|
25
|
da Silva Soares NF, Quagliariello A, Yigitturk S, Martino ME. Gut microbes predominantly act as living beneficial partners rather than raw nutrients. Sci Rep 2023; 13:11981. [PMID: 37488173 PMCID: PMC10366161 DOI: 10.1038/s41598-023-38669-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 07/12/2023] [Indexed: 07/26/2023] Open
Abstract
Animals and their gut microbes mutually benefit their health. Nutrition plays a central role in this, directly influencing both host and microbial fitness and the nature of their interactions. This makes nutritional symbioses a complex and dynamic tri-system of diet-microbiota-host. Despite recent discoveries on this field, full control over the interplay among these partners is challenging and hinders the resolution of fundamental questions, such as how to parse the gut microbes' effect as raw nutrition or as symbiotic partners? To tackle this, we made use of the well-characterized Drosophila melanogaster/Lactiplantibacillus plantarum experimental model of nutritional symbiosis to generate a quantitative framework of gut microbes' effect on the host. By coupling experimental assays and Random Forest analysis, we show that the beneficial effect of L. plantarum strains primarily results from the active relationship as symbionts rather than raw nutrients, regardless of the bacterial strain. Metabolomic analysis of both active and inactive bacterial cells further demonstrated the crucial role of the production of beneficial bacterial metabolites, such as N-acetylated-amino-acids, as result of active bacterial growth and function. Altogether, our results provide a ranking and quantification of the main bacterial features contributing to sustain animal growth. We demonstrate that bacterial activity is the predominant and necessary variable involved in bacteria-mediated benefit, followed by strain-specific properties and the nutritional potential of the bacterial cells. This contributes to elucidate the role of beneficial bacteria and probiotics, creating a broad quantitative framework for host-gut microbiome that can be expanded to other model systems.
Collapse
Affiliation(s)
| | - Andrea Quagliariello
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | - Seren Yigitturk
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
- Food Quality and Design Group, Wageningen University and Research, Wageningen, The Netherlands
| | - Maria Elena Martino
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy.
| |
Collapse
|
26
|
Kaur H, Modgil V, Chaudhary N, Mohan B, Taneja N. Computational Guided Drug Targets Identification against Extended-Spectrum Beta-Lactamase-Producing Multi-Drug Resistant Uropathogenic Escherichia coli. Biomedicines 2023; 11:2028. [PMID: 37509666 PMCID: PMC10377140 DOI: 10.3390/biomedicines11072028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Urinary tract infections (UTIs) are one of the most frequent bacterial infections in the world, both in the hospital and community settings. Uropathogenic Escherichia coli (UPEC) are the predominant etiological agents causing UTIs. Extended-spectrum beta-lactamase (ESBL) production is a prominent mechanism of resistance that hinders the antimicrobial treatment of UTIs caused by UPEC and poses a substantial danger to the arsenal of antibiotics now in use. As bacteria have several methods to counteract the effects of antibiotics, identifying new potential drug targets may help in the design of new antimicrobial agents, and in the control of the rising trend of antimicrobial resistance (AMR). The public availability of the entire genome sequences of humans and many disease-causing organisms has accelerated the hunt for viable therapeutic targets. Using a unique, hierarchical, in silico technique using computational tools, we discovered and described potential therapeutic drug targets against the ESBL-producing UPEC strain NA114. Three different sets of proteins (chokepoint, virulence, and resistance genes) were explored in phase 1. In phase 2, proteins shortlisted from phase 1 were analyzed for their essentiality, non-homology to the human genome, and gut flora. In phase 3, the further shortlisted putative drug targets were qualitatively characterized, including their subcellular location, broad-spectrum potential, and druggability evaluations. We found seven distinct targets for the pathogen that showed no similarity to the human proteome. Thus, possibilities for cross-reactivity between a target-specific antibacterial and human proteins were minimized. The subcellular locations of two targets, ECNA114_0085 and ECNA114_1060, were predicted as cytoplasmic and periplasmic, respectively. These proteins play an important role in bacterial peptidoglycan biosynthesis and inositol phosphate metabolism, and can be used in the design of drugs against these bacteria. Inhibition of these proteins will be helpful to combat infections caused by MDR UPEC.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Vinay Modgil
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Naveen Chaudhary
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Balvinder Mohan
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Neelam Taneja
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
27
|
Carter J, Bettag J, Morfin S, Manithody C, Nagarapu A, Jain A, Nazzal H, Prem S, Unes M, McHale M, Lin CJ, Hutchinson C, Trello G, Jain A, Portz E, Verma A, Swiderska-Syn M, Goldenberg D, Kurashima K. Gut Microbiota Modulation of Short Bowel Syndrome and the Gut-Brain Axis. Nutrients 2023; 15:nu15112581. [PMID: 37299543 DOI: 10.3390/nu15112581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/02/2023] [Accepted: 05/15/2023] [Indexed: 06/12/2023] Open
Abstract
Short bowel syndrome (SBS) is a condition that results from a reduction in the length of the intestine or its functional capacity. SBS patients can have significant side effects and complications, the etiology of which remains ill-defined. Thus, facilitating intestinal adaptation in SBS remains a major research focus. Emerging data supports the role of the gut microbiome in modulating disease progression. There has been ongoing debate on defining a "healthy" gut microbiome, which has led to many studies analyzing the bacterial composition and shifts that occur in gastrointestinal disease states such as SBS and the resulting systemic effects. In SBS, it has also been found that microbial shifts are highly variable and dependent on many factors, including the anatomical location of bowel resection, length, and structure of the remnant bowel, as well as associated small intestinal bacterial overgrowth (SIBO). Recent data also notes a bidirectional communication that occurs between enteric and central nervous systems called the gut-brain axis (GBA), which is regulated by the gut microbes. Ultimately, the role of the microbiome in disease states such as SBS have many clinical implications and warrant further investigation. The focus of this review is to characterize the role of the gut microbiota in short bowel syndrome and its impact on the GBA, as well as the therapeutic potential of altering the microbiome.
Collapse
Affiliation(s)
- Jasmine Carter
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Jeffery Bettag
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Sylvia Morfin
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | | | - Aakash Nagarapu
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Aditya Jain
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Hala Nazzal
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Sai Prem
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Meghan Unes
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Matthew McHale
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Chien-Jung Lin
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Chelsea Hutchinson
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Grace Trello
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Arti Jain
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Edward Portz
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Arun Verma
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | | | - Daniel Goldenberg
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| | - Kento Kurashima
- Department of Pediatrics, Saint Louis University, Saint Louis, MO 63104, USA
| |
Collapse
|
28
|
Mani I. Phage and phage cocktails formulations. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 200:159-169. [PMID: 37739554 DOI: 10.1016/bs.pmbts.2023.04.007] [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: 09/24/2023]
Abstract
Antibiotic-resistant bacterial infection is a major global problem and can be life-threatening. Bacteriophages or phages can be substituted choice over traditional antibiotics treatments. Phages are natural obligate parasites viruses of bacteria, and they can infect and kill antibiotic-sensitive and -resistant bacteria. Further, phages can be utilised as antibacterial agents against various kinds of bacterial infectious diseases. As compared to antibiotics, phages can show a more variety of modes of action and can also be safe in several cases. Phages as a mixture (cocktail) of viral strains are usually used in clinical practices. Generally, to propagate phage cocktails, the individual phage is grown and then mixed to prepare phage cocktails. Antibiotic resistance and biofilm formation can be controlled through formulating phage cocktails that comprise phages infecting single species or by combining phages with non-phages (antibiotics), which may result in a broad spectrum of activity. This chapter briefly highlights the formulations and application of phage cocktails, which are being used to treat various bacterial infections.
Collapse
Affiliation(s)
- Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| |
Collapse
|
29
|
Parra-Llorca A, Pinilla-Gonzlez A, Torrejón-Rodríguez L, Lara-Cantón I, Kuligowski J, Collado MC, Gormaz M, Aguar M, Vento M, Serna E, Cernada M. Effects of Sepsis on Immune Response, Microbiome and Oxidative Metabolism in Preterm Infants. CHILDREN (BASEL, SWITZERLAND) 2023; 10:602. [PMID: 36980160 PMCID: PMC10046958 DOI: 10.3390/children10030602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/03/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023]
Abstract
This is a narrative review about the mechanisms involved in bacterial sepsis in preterm infants, which is an illness with a high incidence, morbidity, and mortality. The role of the innate immune response and its relationship with oxidative stress in the pathogenesis are described as well as their potential implementation as early biomarkers. Moreover, we address the impact that all the mechanisms triggered by sepsis have on the dysbiosis and the changes on neonatal microbiota.
Collapse
Affiliation(s)
- Anna Parra-Llorca
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Alejandro Pinilla-Gonzlez
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Laura Torrejón-Rodríguez
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Inmaculada Lara-Cantón
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Julia Kuligowski
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - María Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), 46980 Valencia, Spain
| | - María Gormaz
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Marta Aguar
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Máximo Vento
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Eva Serna
- Department of Physiology, University of Valencia, 46010 Valencia, Spain
| | - María Cernada
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| |
Collapse
|
30
|
Mazur M, Tomczak H, Łodyga M, Plagens-Rotman K, Merks P, Czarnecka-Operacz M. The Intestinal and Skin Microbiome in Patients with Atopic Dermatitis and Their Influence on the Course of the Disease: A Literature Review. Healthcare (Basel) 2023; 11:healthcare11050766. [PMID: 36900771 PMCID: PMC10001192 DOI: 10.3390/healthcare11050766] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
Bacteria inhabiting the digestive tract are responsible for our health. The microbiome is essential for the development of the immune system and homeostasis of the body. Maintaining homeostasis is very important, but also extremely complicated. The gut microbiome is related to the skin microbiome. It can therefore be assumed that changes in the microbes inhabiting the skin are greatly influenced by the bacteria living in the intestines. Changes in the composition and function of microbes (dysbiosis in the skin and intestines) have recently been linked to changes in the immune response and the development of skin diseases, including atopic dermatitis (AD). This review was compiled by collaborating Dermatologists specializing in atopic dermatitis and psoriasis. A comprehensive review of the current literature was performed using PubMed and limited to relevant case reports and original papers on the skin microbiome in atopic dermatitis. The inclusion criterion was that the paper was published in a peer-reviewed journal in the last 10 years (2012-2022). No limitations on the language of the publication or the type of study were made. It has been shown that any rapid changes in the composition of the microflora may be associated with the appearance of clinical signs and symptoms of the disease. Various studies have proven that the microbiome of many systems (including the intestines) may have a significant impact on the development of the inflammatory process within the skin in the course of AD. It has been shown that an early interaction between the microbiome and immune system may result in a noticeable delay in the onset of atopic diseases. It seems to be of high importance for physicians to understand the role of the microbiome in AD, not only from the pathophysiological standpoint but also in terms of the complex treatment that is required. Perhaps young children diagnosed with AD present specific characteristics of the intestinal microflora. This might be related to the early introduction of antibiotics and dietary manipulations in breastfeeding mothers in the early childhood of AD patients. It is most likely related to the abuse of antibiotics from the first days of life.
Collapse
Affiliation(s)
- Małgorzata Mazur
- College of Health, Beauty Care and Education in Poznań, 60-133 Poznań, Poland
| | - Hanna Tomczak
- Central Microbiological Laboratory, H. Święcicki Clinical Hospital at the Poznan University of Medical Sciences, 60-366 Poznan, Poland
| | - Martha Łodyga
- Department of Medicine Berkshire Medical Center, Pittsfield, MA 01201, USA
| | - Katarzyna Plagens-Rotman
- Center for Pediatric, Adolescent Gynecology and Sexology Division of Gynecology, Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 61-758 Poznan, Poland
- Correspondence:
| | - Piotr Merks
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszyński University, 01-938 Warszawa, Poland
| | - Magdalena Czarnecka-Operacz
- Allergic and Occupational Skin Diseases Unit, Department of Dermatology, Medical University of Poznań, 60-355 Poznan, Poland
| |
Collapse
|
31
|
Advances in Lactobacillus Restoration for β-Lactam Antibiotic-Induced Dysbiosis: A System Review in Intestinal Microbiota and Immune Homeostasis. Microorganisms 2023; 11:microorganisms11010179. [PMID: 36677471 PMCID: PMC9861108 DOI: 10.3390/microorganisms11010179] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023] Open
Abstract
A balanced gut microbiota and their metabolites are necessary for the maintenance of the host's health. The antibiotic-induced dysbiosis can cause the disturbance of the microbial community, influence the immune homeostasis and induce susceptibility to metabolic- or immune-mediated disorders and diseases. The Lactobacillus and their metabolites or components affect the function of the host's immune system and result in microbiota-mediated restoration. Recent data have indicated that, by altering the composition and functions of gut microbiota, antibiotic exposure can also lead to a number of specific pathologies, hence, understanding the potential mechanisms of the interactions between gut microbiota dysbiosis and immunological homeostasis is very important. The Lactobacillus strategies for detecting the associations between the restoration of the relatively imbalanced microbiome and gut diseases are provided in this discussion. In this review, we discuss the recently discovered connections between microbial communities and metabolites in the Lactobacillus treatment of β-lactam antibiotic-induced dysbiosis, and establish the relationship between commensal bacteria and host immunity under this imbalanced homeostasis of the gut microbiota.
Collapse
|
32
|
Shen Y, Yu F, Qiu L, Gao M, Xu P, Zhang L, Liao X, Wang M, Hu X, Sun Y, Pan Y. Ecological influence by colonization of fluoride-resistant Streptococcus mutans in oral biofilm. Front Cell Infect Microbiol 2023; 12:1106392. [PMID: 36699726 PMCID: PMC9868560 DOI: 10.3389/fcimb.2022.1106392] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/14/2022] [Indexed: 01/10/2023] Open
Abstract
Background Dental caries is one of the oldest and most common infections in humans. Improved oral hygiene practices and the presence of fluoride in dentifrices and mouth rinses have greatly reduced the prevalence of dental caries. However, increased fluoride resistance in microbial communities is concerning. Here, we studied the effect of fluoride-resistant Streptococcus mutans (S. mutans) on oral microbial ecology and compare it with wild-type S. mutans in vitro. Methods Biofilm was evaluated for its polysaccharide content, scanning electron microscopy (SEM) imaging, acid-producing ability, and related lactic dehydrogenase (LDH), arginine deiminase (ADS), and urease enzymatic activity determination. Fluorescence in situ hybridization (FISH) and quantitative real-time polymerase chain reaction (qRT-PCR) were used to evaluate the S. mutans ratio within the biofilm. It was followed by 16S rRNA sequencing to define the oral microbial community. Results Fluoride-resistant S. mutans produced increased polysaccharides in presence of NaF (P < 0.05). The enzymatic activities related to both acid and base generation were less affected by the fluoride. In presence of 275 ppm NaF, the pH in the fluoride-resistant strain sample was lower than the wild type. We observed that with the biofilm development and accumulative fluoride concentration, the fluoride-resistant strain had positive relationships with other bacteria within the oral microbial community, which enhanced its colonization and survival. Compared to the wild type, fluoride-resistant strain significantly increased the diversity and difference of oral microbial community at the initial stage of biofilm formation (4 and 24 h) and at a low fluoride environment (0 and 275 ppm NaF) (P < 0.05). Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that fluoride-resistant strain enhanced the metabolic pathways and glucose transfer. Conclusions Fluoride-resistant S. mutans affected the microecological balance of oral biofilm and its cariogenic properties in vitro, indicating its negative impact on fluoride's caries prevention effect.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yan Sun
- *Correspondence: Yihuai Pan, ; Yan Sun,
| | | |
Collapse
|
33
|
Sheng M, Xu S, Chen WW, Li FQ, Zhong YM, Ouyang YX, Liao YL, Lai P. A bibliometric analysis of studies on the gut microbiota in cardiovascular disease from 2004 to 2022. Front Cell Infect Microbiol 2023; 12:1083995. [PMID: 36683688 PMCID: PMC9852829 DOI: 10.3389/fcimb.2022.1083995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/06/2022] [Indexed: 01/09/2023] Open
Abstract
Background Increasing evidence indicates that the gut microbiota (GM) is linked to cardiovascular disease (CVD). Many studies on the GM in CVD have been published in the last decade. However, bibliometric analysis in this field is still lacking. Methods On 30 September 2022, a search of the Web of Science™ (WoS; Clarivate™, Philadelphia, PA, USA) yielded 1,500 articles and reviews on the GM and CVD. Microsoft Excel and CiteSpace and VOSviewer software were used to analyze publication trends and research hotspots in this field. Results Our search generated 1,708 publications on the GM in CVD published between 2004 and 2022, and 1,500 articles and review papers were included in the final analysis. The number of publications relating to the GM in CVD increased from 1 in 2004 to 350 in 2021. China (485 publications, 9,728 non-self-citations, and an H-index of 47) and the USA (418 publications, 24,918 non-self-citations, and an H-index of 82) contributed 32.31%, and 27.85%, respectively, of the total number of publications. Examination of the number of publications (Np) and number of citations, excluding self-citations (Nc), of individual authors showed that Y. L. Tian (Np: 18, Nc: 262, and H-index: 12), from China, is the most productive author, followed by R. Knight (Np: 16, Nc: 3,036, and H-index: 15) and M. Nieuwdorp (Np: 16, Nc: 503, and H-index: 9). The Chinese Academy of Medical Sciences and Peking Union Medical College accounted for the largest number of publications (Np: 62, Nc: 3,727, and H-index: 13, average citation number (ACN): 60.11). The journal Nutrients had the most publications (Np: 73, Nc: 2,036, and ACN: 27.89). The emerging keywords in this field were "monooxygenase 3" (strength 3.24, 2020-2022), "short-chain fatty acid" (strength 4.63, 2021-2022), "fatty liver disease" (strength 3.18, 2021-2022), "metabolic disease" (strength 3.04, 2021-2022), "Mediterranean diet" (strength 2.95, 2021-2022), "prevention" (strength 2.77, 2021-2022), and "intestinal barrier" (strength 2.8, 2021-2022). Conclusion Publications on the GM in CVD rapidly increased in the last decade. The USA was the most influential country in publications in this field, followed by China. The journal with the most publications was Nutrients. Monooxygenase-3, short-chain fatty acids, fatty liver disease, metabolic disease, the Mediterranean diet, intestinal barrier, and prevention are the current hotspots or potential hotspots for future study.
Collapse
Affiliation(s)
- Ming Sheng
- Department of Library, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Shuquan Xu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Wei-Wei Chen
- Department of Pharmacology, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Fa-Quan Li
- Department of Cardiology, The First Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yi-Ming Zhong
- Department of Cardiology, The First Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yi-Xiang Ouyang
- Department of Library, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yong-Ling Liao
- Department of Cardiology, The First Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Ping Lai
- Department of Cardiology, The First Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|
34
|
Du L, Chen B, Cheng F, Kim J, Kim JJ. Effects of Helicobacter pylori Therapy on Gut Microbiota: A Systematic Review and Meta-Analysis. Dig Dis 2022; 42:102-112. [PMID: 36228588 DOI: 10.1159/000527047] [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: 12/11/2021] [Accepted: 09/04/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Although indications for evaluation and treatment of Helicobacter pylori infection are broadening to include primary prevention for gastric adenocarcinoma, potential adverse effects on gut microbiota have been raised. We performed a systematic review and meta-analysis to evaluate the effects of H. pylori therapy on gut microbiota. METHODS PubMed, EMBASE, Cochrane Library, and Web of Science (to 4/2021) were searched for studies quantitatively evaluating microbiota before and after H. pylori therapy. Meta-analysis was performed to assess early (<1 year) and long-term (≥1 year) effects on gut microbiota after H. pylori treatment. Subgroup analysis evaluating the effects of H. pylori therapy with addition of probiotics on gut microbiota was also performed. RESULTS Thirty studies (N = 1,218) met the criteria. Early after H. pylori therapy, intestinal microbial diversity was reduced in nearly all studies. At the genus level, reduction in the abundance of Enterococcus, while increase in Lactobacillus, Bifidobacterium, and Bacteroides counts were observed. However, Enterococcus, Lactobacillus, Bifidobacterium, and Bacteroides counts remained stable in patients who received probiotics with H. pylori therapy. At the phylum level, the relative abundance of Actinobacteria and Firmicutes increased after treatment. At ≥1 year, intestinal microbial diversity normalized in six of seven studies. No differences in the relative abundance of Actinobacteria, Firmicute, Bacteroidetes, and Proteobacteria were observed ≥1 year after therapy. CONCLUSION The impact of H. pylori therapy on gut microbiota appears transient with early changes largely resolving after 1 year. Probiotics may reduce the early impact of H. pylori therapy on gut microbiota.
Collapse
Affiliation(s)
- Lijun Du
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China,
| | - Binrui Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fangli Cheng
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jeffrey Kim
- Department of Family Medicine, Loma Linda University Health, Loma Linda, California, USA
| | - John J Kim
- Division of Gastroenterology, Loma Linda University Health, Loma Linda, California, USA
| |
Collapse
|
35
|
Yakovlev SV, Suvorova MP. Rationale for choosing an antibiotic for the treatment of cystitis: recommendations of clinical pharmacologists: A review. TERAPEVT ARKH 2022; 94:1006-1013. [DOI: 10.26442/00403660.2022.08.201775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022]
Abstract
In recent years, the harmonization of domestic and foreign clinical recommendations for the treatment of cystitis has been achieved. Nitrofurans and fosfomycin trometamol are recommended as first line therapy antibiotics, and oral 3rd generation of cephalosporins are recommended as alternative antibiotics; fluoroquinolones are excluded from the recommended medications due to an unfavorable safety profile. The main rationale for inclusion of antibiotics in the recommendations as a first line therapy of cystitis is the level of resistance of uropathogens to antibiotics, primarily Escherichia coli. Stable low level of resistance of E. coli in Russia was noted to nitrofurans and fosfomycin (5%), higher to cephalosporins. Among nitrofurans, furazidine is characterized by higher activity against E. coli compared to nitrofurantoin. The potassium salt of furazidine in dosage form with magnesium carbonate is preferred, since it is characterized by higher bioavailability and provides a therapeutic level of concentrations in urine above the MIC during the entire dosing period. Due to the global increase in the resistance of uropathogens observed in recent years, experts have begun to pay more and more attention to the ecological safety of antimicrobial therapy in order to minimize the risk of concomitant (collateral) damage, contributing to the selection of multi-drug resistant strains of microorganisms. In the latest WHO document of 2021, experts divided antibiotics into three groups (ACCESS, WATCH, RESERVE) according to the priority of choice. The ACCESS group of drugs for the treatment of cystitis includes nitrofurantoin and furazidine as agents with minimal collateral effect, while fosfomycin trometamol and cephalosporins are listed in the WATCH group. Thus, from the standpoint of ecological safety, WHO experts recommend prescribing nitrofurans in the treatment of cystitis in the first line of therapy.
Collapse
|
36
|
Xu HM, Xu WM, Zhang L. Current Status of Phage Therapy against Infectious Diseases and Potential Application beyond Infectious Diseases. Int J Clin Pract 2022; 2022:4913146. [PMID: 36263241 PMCID: PMC9550513 DOI: 10.1155/2022/4913146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/19/2022] [Indexed: 12/02/2022] Open
Abstract
Intestinal microbiota plays a key role in regulating the pathogenesis of human disease and maintaining health. Many diseases, mainly induced by bacteria, are on the rise due to the emergence of antibiotic-resistant strains. Intestinal microorganisms include organisms such as bacteria, viruses, and fungi. They play an important role in maintaining human health. Among these microorganisms, phages are the main members of intestinal viromes. In particular, the viral fraction, composed essentially of phages, affects homeostasis by exerting selective pressure on bacterial communities living in the intestinal tract. In recent years, with the widespread use and even abuse of antibacterial drugs, more and more drug-resistant bacteria have been found, and they show a trend of high drug resistance and multidrug resistance. Therefore, it has also become increasingly difficult to treat serious bacterial infections. Phages, a natural antibacterial agent with strong specificity and rapid proliferation, have come back to the field of vision of clinicians and scholars. In this study, the current state of research on intestinal phages was discussed, with an exploration of the impact of phage therapy against infectious diseases, as well as potential application beyond infectious diseases.
Collapse
Affiliation(s)
- Hao-Ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Wen-Min Xu
- Department of Endoscopy, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510091, China
| | - Long Zhang
- Department of Endoscopy, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510091, China
| |
Collapse
|
37
|
Gut microbiota: a new avenue to reveal pathological mechanisms of constipation. Appl Microbiol Biotechnol 2022; 106:6899-6913. [PMID: 36190540 DOI: 10.1007/s00253-022-12197-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022]
Abstract
Constipation is very pervasive all over the world. It is a common multifactorial gastrointestinal disease, and its etiology and pathomechanism are not completely clear. Now, increasing evidence shows that intestinal flora is closely related to constipation. Intestinal flora is the largest microbiota in the human body and has powerful metabolic functions. Intestinal flora can produce a variety of metabolites, such as bile acids, short-chain fatty acids, tryptophan metabolites, and methane, which have important effects on intestinal motility and secretion. The host can also monitor the intestinal flora and regulate gut dysbacteriosis in constipation. To explore the relationship between intestinal flora and host, the combination of multiomics technology has become the powerful and effective method. Furthermore, the homeostasis restoration of intestinal flora also provides a new strategy for the treatment of constipation. This review aims to explore the interaction between intestinal flora and host in constipation, which contributes to disclose the pathogenesis of constipation and the development of novel drugs for the treatment of constipation from the perspective of intestinal flora. KEY POINTS: • This review highlights the regulation of gut microbiota on the intestinal motility and secretion of host. • The current review gives an insight into the role of the host on the recognition and regulation of intestinal ecology under constipation. • The article also introduces some novel methods of current gut microbiota research and gut microbiota-based constipation therapies.
Collapse
|
38
|
Duszka K. Versatile Triad Alliance: Bile Acid, Taurine and Microbiota. Cells 2022; 11:2337. [PMID: 35954180 PMCID: PMC9367564 DOI: 10.3390/cells11152337] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/21/2022] [Accepted: 07/24/2022] [Indexed: 11/21/2022] Open
Abstract
Taurine is the most abundant free amino acid in the body, and is mainly derived from the diet, but can also be produced endogenously from cysteine. It plays multiple essential roles in the body, including development, energy production, osmoregulation, prevention of oxidative stress, and inflammation. Taurine is also crucial as a molecule used to conjugate bile acids (BAs). In the gastrointestinal tract, BAs deconjugation by enteric bacteria results in high levels of unconjugated BAs and free taurine. Depending on conjugation status and other bacterial modifications, BAs constitute a pool of related but highly diverse molecules, each with different properties concerning solubility and toxicity, capacity to activate or inhibit receptors of BAs, and direct and indirect impact on microbiota and the host, whereas free taurine has a largely protective impact on the host, serves as a source of energy for microbiota, regulates bacterial colonization and defends from pathogens. Several remarkable examples of the interaction between taurine and gut microbiota have recently been described. This review will introduce the necessary background information and lay out the latest discoveries in the interaction of the co-reliant triad of BAs, taurine, and microbiota.
Collapse
Affiliation(s)
- Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
39
|
Boudar Z, Sehli S, El Janahi S, Al Idrissi N, Hamdi S, Dini N, Brim H, Amzazi S, Nejjari C, Lloyd-Puryear M, Ghazal H. Metagenomics Approaches to Investigate the Neonatal Gut Microbiome. Front Pediatr 2022; 10:886627. [PMID: 35799697 PMCID: PMC9253679 DOI: 10.3389/fped.2022.886627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/23/2022] [Indexed: 12/03/2022] Open
Abstract
Early infancy is critical for the development of an infant's gut flora. Many factors can influence microbiota development during the pre- and postnatal periods, including maternal factors, antibiotic exposure, mode of delivery, dietary patterns, and feeding type. Therefore, investigating the connection between these variables and host and microbiome interactions in neonatal development would be of great interest. As the "unculturable" era of microbiome research gives way to an intrinsically multidisciplinary field, microbiome research has reaped the advantages of technological advancements in next-generation sequencing, particularly 16S rRNA gene amplicon and shotgun sequencing, which have considerably expanded our knowledge about gut microbiota development during early life. Using omics approaches to explore the neonatal microbiome may help to better understand the link between the microbiome and newborn diseases. Herein, we summarized the metagenomics methods and tools used to advance knowledge on the neonatal microbiome origin and evolution and how the microbiome shapes early and late individuals' lives for health and disease. The way to overcome limitations in neonatal microbiome studies will be discussed.
Collapse
Affiliation(s)
- Zakia Boudar
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Sofia Sehli
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Sara El Janahi
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Najib Al Idrissi
- Department of Surgery, Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Salsabil Hamdi
- Laboratory of Genomics and Bioinformatics, School of Pharmacy, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Nouzha Dini
- Mother and Child Department, Cheikh Khalifa International University Hospital, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Hassan Brim
- Department of Pathology, Howard University, Washington, DC, United States
| | - Saaïd Amzazi
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, and Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Chakib Nejjari
- Department of Epidemiology and Biostatistics, International School of Public Health, Mohammed VI University of Health Sciences, Casablanca, Morocco
- Department of Epidemiology and Public Health, Faculty of Medicine, University Sidi Mohammed Ben Abdellah, Fez, Morocco
| | | | - Hassan Ghazal
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
- National Center for Scientific and Technical Research, Rabat, Morocco
| |
Collapse
|
40
|
Liu Y, Liu C, Wu H, Meng Q, Zhou Z. Small Intestine Microbiome and Metabolome of High and Low Residual Feed Intake Angus Heifers. Front Microbiol 2022; 13:862151. [PMID: 35531283 PMCID: PMC9069012 DOI: 10.3389/fmicb.2022.862151] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/17/2022] [Indexed: 12/02/2022] Open
Abstract
The gastrointestinal tract (GIT) contains complex microbial communities and plays an essential role in the overall health of the host. Previous studies of beef cattle feed efficiency have primarily concentrated on the ruminal microbiota because it plays a key role in energy production and nutrient supply in the host. Although the small intestine is the important site of post-ruminal digestion and absorption of nutrients, only a few studies have explored the relationship between the microbial populations in the small intestine and feed efficiency. Moreover, variations in GIT metabolites contribute to differences in feed efficiency. The objective of this study was to investigate relationships among bacterial populations of duodenum, jejunum, ileum; microbial metabolites; and RFI phenotype of beef cattle. We carried out by using Illumina MiSeq sequencing of the 16S rRNA V3-V4 region and liquid chromatography-mass spectrometry (LC–MS). In the duodenum, the relative abundances of Firmicutes ( p < 0.01), Lachnospiraceae, Ruminococcaceae, Family_XIII, Christensenellaceae, Christensenellaceae_R-7_group ( p < 0.05), and Lachnospiraceae_NK3A20_group ( p < 0.05) were higher in the low residual feed intake (LRFI) group compared with the high residual feed intake (HRFI) group, whereas the HRFI group had higher abundances of Proteobacteria and Acinetobacter ( p < 0.01). In the jejunum, the relative abundances of Lachnospiraceae and Lachnospiraceae_NK3A20_group were higher in the LRFI group ( p < 0.05). In the ileum, the relative abundances of Ruminococcaceae ( p < 0.01), Christensenellaceae, Christensenellaceae_R-7_group, and Ruminococcus_2 were also higher in the LRFI group ( p < 0.05). Moreover, the genera Lachnospiraceae_NK3A20_group, Christensenellaceae_R-7_group, and Ruminococcus_2 were negatively associated with RFI, while the genus Acinetobacter was positively associated with RFI. The metabolomics analysis revealed that the LRFI group significantly improved protein digestion and absorption, as well as glycerophospholipid metabolism in the duodenum, jejunum, ileum. The correlation between intestinal microorganisms and metabolites revealed that some microorganisms play an important role in amino acid metabolism, glycerophospholipid metabolism, nutrient digestion and absorption, and antioxidant enhancement. The present study provides a better understanding of the small intestinal microbiota and metabolites of beef cattle with different RFI phenotypes and the relationships among them, which are potentially important for the improvement of beef cattle feed efficiency.
Collapse
Affiliation(s)
- Yue Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chang Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Hao Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qingxiang Meng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhenming Zhou
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
41
|
Bornbusch SL, Greene LK, Rahobilalaina S, Calkins S, Rothman RS, Clarke TA, LaFleur M, Drea CM. Gut microbiota of ring-tailed lemurs (Lemur catta) vary across natural and captive populations and correlate with environmental microbiota. Anim Microbiome 2022; 4:29. [PMID: 35484581 PMCID: PMC9052671 DOI: 10.1186/s42523-022-00176-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 03/29/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Inter-population variation in host-associated microbiota reflects differences in the hosts' environments, but this characterization is typically based on studies comparing few populations. The diversity of natural habitats and captivity conditions occupied by any given host species has not been captured in these comparisons. Moreover, intraspecific variation in gut microbiota, generally attributed to diet, may also stem from differential acquisition of environmental microbes-an understudied mechanism by which host microbiomes are directly shaped by environmental microbes. To more comprehensively characterize gut microbiota in an ecologically flexible host, the ring-tailed lemur (Lemur catta; n = 209), while also investigating the role of environmental acquisition, we used 16S rRNA sequencing of lemur gut and soil microbiota sampled from up to 13 settings, eight in the wilderness of Madagascar and five in captivity in Madagascar or the U.S. Based on matched fecal and soil samples, we used microbial source tracking to examine covariation between the two types of consortia. RESULTS The diversity of lemur gut microbes varied markedly within and between settings. Microbial diversity was not consistently greater in wild than in captive lemurs, indicating that this metric is not necessarily an indicator of host habitat or environmental condition. Variation in microbial composition was inconsistent both with a single, representative gut community for wild conspecifics and with a universal 'signal of captivity' that homogenizes the gut consortia of captive animals. Despite the similar, commercial diets of captive lemurs on both continents, lemur gut microbiomes within Madagascar were compositionally most similar, suggesting that non-dietary factors govern some of the variability. In particular, soil microbial communities varied across geographic locations, with the few samples from different continents being the most distinct, and there was significant and context-specific covariation between gut and soil microbiota. CONCLUSIONS As one of the broadest, single-species investigations of primate microbiota, our study highlights that gut consortia are sensitive to multiple scales of environmental differences. This finding begs a reevaluation of the simple 'captive vs. wild' dichotomy. Beyond the important implications for animal care, health, and conservation, our finding that environmental acquisition may mediate aspects of host-associated consortia further expands the framework for how host-associated and environmental microbes interact across different microbial landscapes.
Collapse
Affiliation(s)
- Sally L. Bornbusch
- Department of Evolutionary Anthropology, Duke University, Durham, NC USA
| | | | | | - Samantha Calkins
- Department of Psychology, Program in Animal Behavior and Conservation, Hunter College, New York, NY USA
| | - Ryan S. Rothman
- Institute for the Conservation of Tropical Environments, Interdepartmental Doctoral Program in Anthropological Sciences, Stony Brook University, Stony Brook, NY USA
| | - Tara A. Clarke
- Department of Sociology and Anthropology, North Carolina State University, Raleigh, NC USA
| | - Marni LaFleur
- Department of Anthropology, University of San Diego, 5998 Alcala Park, San Diego, CA USA
| | - Christine M. Drea
- Department of Evolutionary Anthropology, Duke University, Durham, NC USA
| |
Collapse
|
42
|
Evaluation of Host Depletion and Extraction Methods for Shotgun Metagenomic Analysis of Bovine Vaginal Samples. Microbiol Spectr 2022; 10:e0041221. [PMID: 35404108 PMCID: PMC9045270 DOI: 10.1128/spectrum.00412-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The reproductive tract metagenome plays a significant role in the various reproductive system functions, including reproductive cycles, health, and fertility. One of the major challenges in bovine vaginal metagenome studies is host DNA contamination, which limits the sequencing capacity for metagenomic content and reduces the accuracy of untargeted shotgun metagenomic profiling. This is the first study comparing the effectiveness of different host depletion and DNA extraction methods for bovine vaginal metagenomic samples. The host depletion methods evaluated were slow centrifugation (Soft-spin), NEBNext Microbiome DNA Enrichment kit (NEBNext), and propidium monoazide (PMA) treatment, while the extraction methods were DNeasy Blood and Tissue extraction (DNeasy) and QIAamp DNA Microbiome extraction (QIAamp). Soft-spin and QIAamp were the most effective host depletion method and extraction methods, respectively, in reducing the number of cattle genomic content in bovine vaginal samples. The reduced host-to-microbe ratio in the extracted DNA increased the sequencing depth for microbial reads in untargeted shotgun sequencing. Bovine vaginal samples extracted with QIAamp presented taxonomical profiles which closely resembled the mock microbial composition, especially for the recovery of Gram-positive bacteria. Additionally, samples extracted with QIAamp presented extensive functional profiles with deep coverage. Overall, a combination of Soft-spin and QIAamp provided the most robust representation of the vaginal microbial community in cattle while minimizing host DNA contamination. IMPORTANCE In addition to the host tissue collected during the sampling process, bovine vaginal samples are saturated with large amounts of extracellular DNA and secreted proteins that are essential for physiological purposes, including the reproductive cycle and immune defense. Due to the high host-to-microbe genome ratio, which hampers the sequencing efficacy for metagenome samples and the recovery of the actual metagenomic profiles, bovine vaginal samples cannot benefit from the full potential of shotgun sequencing. This is the first investigation on the most effective host depletion and extraction methods for bovine vaginal metagenomic samples. This study demonstrated an effective combination of host depletion and extraction methods, which harvested higher percentages of 16S rRNA genes and microbial reads, which subsequently led to a taxonomical profile that resembled the actual community and a functional profile with deeper coverage. A representative metagenomic profile is essential for investigating the role of the bovine vaginal metagenome for both reproductive function and susceptibility to infections.
Collapse
|
43
|
Mohd Salleh MH, Esa Y, Ngalimat MS, Chen PN. Faecal DNA metabarcoding reveals novel bacterial community patterns of critically endangered Southern River Terrapin, Batagur affinis. PeerJ 2022; 10:e12970. [PMID: 35368336 PMCID: PMC8973471 DOI: 10.7717/peerj.12970] [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: 10/04/2021] [Accepted: 01/30/2022] [Indexed: 01/11/2023] Open
Abstract
Southern River Terrapin, Batagur affinis, is a freshwater turtle listed as critically endangered on the IUCN Red List since 2000. Many studies suggest that faecal DNA metabarcoding can shield light on the host-associated microbial communities that play important roles in host health. Thus, this study aimed to characterise and compare the faecal bacterial community between captive and wild B. affinis using metabarcoding approaches. A total of seven faeces samples were collected from captive (N = 5) and wild (N = 2) adult B. affinis aseptically, crossing the East and West coast of peninsular Malaysia. The DNA was extracted from the faeces samples, and the 16S rRNA gene (V3-V4 region) was amplified using polymerase chain reaction (PCR). The amplicon was further analysed using SILVA and DADA2 pipelines. In total, 297 bacterial communities taxonomic profile (phylum to genus) were determined. Three phyla were found in high abundance in all faeces samples, namely Firmicutes (38.69%), Bacteroidetes (24.52%), and Fusobacteria (6.95%). Proteobacteria were detected in all faeces samples (39.63%), except the wild sample, KBW3. Under genus level, Cetobacteriumwas found as the most abundant genus (67.79%), followed by Bacteroides (24.56%) and Parabacteroides (21.78%). The uncultured genus had the highest abundance (88.51%) even though not detected in the BK31 and KBW2 samples. The potential probiotic genera (75.00%) were discovered to be more dominant in B. affinis faeces samples. Results demonstrated that the captive B. affinis faeces samples have a greater bacterial variety and richness than wild B. affinis faeces samples. This study has established a starting point for future investigation of the gut microbiota of B. affinis.
Collapse
Affiliation(s)
- Mohd Hairul Mohd Salleh
- Department of Aquaculture, Faculty of Agriculture, Universiti Putra Malaysia, Serdang, Selangor, Malaysia,Royal Malaysian Customs Department, Presint 2, Putrajaya, Malaysia
| | - Yuzine Esa
- Department of Aquaculture, Faculty of Agriculture, Universiti Putra Malaysia, Serdang, Selangor, Malaysia,International Institute of Aquaculture and Aquatic Sciences, Universiti Putra Malaysia, Port Dickson, Negeri Sembilan, Malaysia
| | - Mohamad Syazwan Ngalimat
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Pelf Nyok Chen
- Turtle Conservation Society of Malaysia, Kemaman, Terengganu, Malaysia
| |
Collapse
|
44
|
Morshedzadeh N, Rahimlou M, Shahrokh S, Mirmiran P, Zali MR. Nutritional management of inflammatory bowel disease; an overview of the evidences. Diabetes Metab Syndr 2022; 16:102440. [PMID: 35247656 DOI: 10.1016/j.dsx.2022.102440] [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: 09/01/2021] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND AIMS Inflammatory bowel disease (IBD) is a chronic relapsing-remitting systemic disease and one of the most common gastrointestinal diseases that affect many people. This review designed to report the latest findings on the association between some nutrients and IBD. METHODS A review was performed to summarize the effect of various aspects of nutrition and diet on clinical course, the severity of disease, intestinal epithelial inflammation, inflammatory and oxidative stress markers. Literature searches were conducted in PubMed and Google Scholar up to June 27, 2021. RESULTS Various studies have shown that an unhealthy diet and deficiency of some nutrients are involved in the etiology of IBD. It has also been shown that intestinal dysbiosis can increase the risk of developing IBD. The results of some studies have shown that supplementation with some nutrients such as omega-3 polyunsaturated fatty acids and vitamin D and probiotics may have beneficial results in patients with IBD. Adherence to some restrictive diets has also been helpful in some studies. CONCLUSIONS Following proper nutritional approaches can play an essential role in managing IBD symptoms. Further studies are needed to substantiate some of these findings.
Collapse
Affiliation(s)
- Nava Morshedzadeh
- Department of Nutrition, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehran Rahimlou
- Department of Nutrition, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Shabnam Shahrokh
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
The Nutrition-Microbiota-Physical Activity Triad: An Inspiring New Concept for Health and Sports Performance. Nutrients 2022; 14:nu14050924. [PMID: 35267899 PMCID: PMC8912693 DOI: 10.3390/nu14050924] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
The human gut microbiota is currently the focus of converging interest in many diseases and sports performance. This review presents gut microbiota as a real “orchestra conductor” in the host’s physio(patho)logy due to its implications in many aspects of health and disease. Reciprocally, gut microbiota composition and activity are influenced by many different factors, such as diet and physical activity. Literature data have shown that macro- and micro-nutrients influence gut microbiota composition. Cumulative data indicate that gut bacteria are sensitive to modulation by physical activity, as shown by studies using training and hypoactivity models. Sports performance studies have also presented interesting and promising results. Therefore, gut microbiota could be considered a “pivotal” organ for health and sports performance, leading to a new concept: the nutrition-microbiota-physical activity triad. The next challenge for the scientific and medical communities is to test this concept in clinical studies. The long-term aim is to find the best combination of the three elements of this triad to optimize treatments, delay disease onset, or enhance sports performance. The many possibilities offered by biotic supplementation and training modalities open different avenues for future research.
Collapse
|
46
|
Bozzi Cionci N, Lucaccioni L, Pietrella E, Ficara M, Spada C, Torelli P, Bedetti L, Lugli L, Di Gioia D, Berardi A. Antibiotic Exposure, Common Morbidities and Main Intestinal Microbial Groups in Very Preterm Neonates: A Pilot Study. Antibiotics (Basel) 2022; 11:237. [PMID: 35203839 PMCID: PMC8868158 DOI: 10.3390/antibiotics11020237] [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: 12/22/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 01/27/2023] Open
Abstract
Prematurity exposes newborns to increased risks of infections and it is associated with critical morbidities. Preterm infants often require antibiotic therapies that can affect the correct establishment of gut microbiota. The aim of this study was to investigate targeted intestinal bacteria in preterm neonates with common morbidities and receiving antibiotic treatments of variable duration. Stool samples were collected after birth, at 15, 30 and 90 days of life. qPCR quantification of selected microbial groups (Bifidobacterium spp., Bacteroides fragilis group, Enterobacteriaceae, Clostridium cluster I and total bacteria) was performed and correlation between their levels, the duration of antibiotic treatment and different clinical conditions was studied. An increasing trend over time was observed for all microbial groups, especially for Bifdobacterium spp. Prolonged exposure to antibiotics in the first weeks of life affected Clostridium and B. fragilis levels, but these changes no longer persisted at 90 days of life. Variations of bacterial counts were associated with the length of hospital stay, feeding and mechanical ventilation. Late-onset sepsis and patent ductus arteriosus reduced the counts of Bifidobacterium, whereas B. fragilis was influenced by compromised respiratory conditions. This study can be a start point for the identification of microbial biomarkers associated with some common morbidities and tailored strategies for a healthy microbial development.
Collapse
Affiliation(s)
- Nicole Bozzi Cionci
- Department of Agricultural and Food Sciences, University of Bologna, 40127 Bologna, Italy; (N.B.C.); (D.D.G.)
| | - Laura Lucaccioni
- Pediatric Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Elisa Pietrella
- Pediatric Unit, Department of Medical for Mothers and Children, Ramazzini Hospital, 41012 Carpi, Italy;
| | - Monica Ficara
- Pediatric Unit, Department of Medical for Mothers and Children, Bufalini Hospital, 47521 Cesena, Italy;
| | - Caterina Spada
- Neonatal Intensive Care Unit, Department of Medical for Mothers and Children, Bufalini Hospital, 47521 Cesena, Italy;
| | - Paola Torelli
- Neonatal Intensive Care Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (P.T.); (L.B.); (L.L.); (A.B.)
| | - Luca Bedetti
- Neonatal Intensive Care Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (P.T.); (L.B.); (L.L.); (A.B.)
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Licia Lugli
- Neonatal Intensive Care Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (P.T.); (L.B.); (L.L.); (A.B.)
| | - Diana Di Gioia
- Department of Agricultural and Food Sciences, University of Bologna, 40127 Bologna, Italy; (N.B.C.); (D.D.G.)
| | - Alberto Berardi
- Neonatal Intensive Care Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (P.T.); (L.B.); (L.L.); (A.B.)
| |
Collapse
|
47
|
Tikunov AY, Shvalov AN, Morozov VV, Babkin IV, Seledtsova GV, Voloshina IO, Ivanova IP, Bardasheva AV, Morozova VV, Vlasov VV, Tikunova NV. Taxonomic composition and biodiversity of the gut microbiome from patients with irritable bowel syndrome, ulcerative colitis, and asthma. Vavilovskii Zhurnal Genet Selektsii 2022; 25:864-873. [PMID: 35083405 PMCID: PMC8753531 DOI: 10.18699/vj21.100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 02/19/2021] [Accepted: 03/17/2021] [Indexed: 11/19/2022] Open
Abstract
To date, the association of an imbalance of the intestinal microbiota with various human diseases, including both diseases of the gastrointestinal tract and disorders of the immune system, has been shown. However, despite the huge amount of accumulated data, many key questions still remain unanswered. Given limited data on the composition of the gut microbiota in patients with ulcerative colitis (UC) and irritable bowel syndrome (IBS) from different parts of Siberia, as well as the lack of data on the gut microbiota of patients with bronchial asthma (BA), the aim of the study was to assess the biodiversity of the gut microbiota of patients with IBS, UC and BA in comparison with those of healthy volunteers (HV). In this study, a comparative assessment of the biodiversity and taxonomic structure of gut microbiome was conducted based on the sequencing of 16S rRNA genes obtained from fecal samples of patients with IBS, UC, BA and volunteers. Sequences of the Firmicutes and Bacteroidetes types dominated in all samples studied. The third most common in all samples were sequences of the Proteobacteria type, which contains pathogenic and opportunistic bacteria. Sequences of the Actinobacteria type were, on average, the fourth most common. The results showed the presence of dysbiosis in the samples from patients compared to the sample from HVs. The ratio of Firmicutes/Bacteroidetes was lower in the IBS and UC samples than in HV and higher the BA samples. In the samples from patients with intestinal diseases (IBS and UC), an increase in the proportion of sequences of the Bacteroidetes type and a decrease in the proportion of sequences of the Clostridia class, as well as the Ruminococcaceae, but not Erysipelotrichaceae family, were found. The IBS, UC, and BA samples had signif icantly more Proteobacteria sequences, including Methylobacterium, Sphingomonas, Parasutterella, Halomonas, Vibrio, as well as Escherichia spp. and Shigella spp. In the gut microbiota of adults with BA, a decrease in the proportion of Roseburia, Lachnospira, Veillonella sequences was detected, but the share of Faecalibacterium and Lactobacillus sequences was the same as in healthy individuals. A signif icant increase in the proportion of Halomonas and Vibrio sequences in the gut microbiota in patients with BA has been described for the f irst time.
Collapse
Affiliation(s)
- A. Y. Tikunov
- Institute of Сhemical Biology аnd Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences
| | - A. N. Shvalov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor
| | - V. V. Morozov
- Institute of Сhemical Biology аnd Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences
| | - I. V. Babkin
- Institute of Сhemical Biology аnd Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences
| | | | - I. O. Voloshina
- Institute of Сhemical Biology аnd Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences
| | | | - A. V. Bardasheva
- Institute of Сhemical Biology аnd Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences
| | - V. V. Morozova
- Institute of Сhemical Biology аnd Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences
| | - V. V. Vlasov
- Institute of Сhemical Biology аnd Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences
| | - N. V. Tikunova
- Institute of Сhemical Biology аnd Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences
| |
Collapse
|
48
|
Gut microbiome and type 2 diabetes. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 191:175-185. [DOI: 10.1016/bs.pmbts.2022.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
49
|
Zhang Y, Zhang P, Li Y. Gut microbiota-mediated ferroptosis contributes to mercury exposure-induced brain injury in common carp. Metallomics 2021; 14:6461106. [PMID: 34905050 DOI: 10.1093/mtomcs/mfab072] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/30/2021] [Indexed: 11/14/2022]
Abstract
Mercury is a heavy metal which causes irreversible toxicity to fish and is found in aquatic environments around the world. The purpose of this study was to investigate the relative mechanism of mercury exposure on brain injury in common carp. The results showed that mercury exposure could induce brain injury and memory loss in common carp. Meanwhile, mercury exposure could induce neuronal ferroptosis. The ferroptosis inhibitor ferrostatin-1 attenuated mercury-induced brain injury. However, in an vitro study, mercury did not induce ferroptosis, and ferrostatin-1 did not attenuate mercury-induced common carp brain cell death. Therefore, we speculated that mercury exposure-induced ferroptosis might occur through other pathways. Studies have shown that the gut microbiota contributes to the pathological process of heavy metal-induced injury. Therefore, we detected the effects of mercury exposure on the gut microbiota composition. The results showed that the composition and diversity of the gut microbiota were affected by mercury chloride. Surprisingly, we found that the abundance of Aeromonas, one of the most important pathogenic bacteria of fish, increased significantly. Subsequently, we isolated Aeromonas hydrophila from mercury-exposed carp and this bacteria could lead to brain injury and ferroptosis in common carp. These results suggested that mercury exposure-induced brain injury partly by increasing intestinal Aeromonas hydrophila, which led to ferroptosis in common carp.
Collapse
Affiliation(s)
- Yue Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.,Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, 130118, China
| | - Peijun Zhang
- Health Monitoring and Inspection Center of Jilin Province, Changchun, 130062, China
| | - Yuehong Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.,Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, 130118, China
| |
Collapse
|
50
|
Schade L, Mesa D, Faria AR, Santamaria JR, Xavier CA, Ribeiro D, Hajar FN, Azevedo VF. The gut microbiota profile in psoriasis: a Brazilian case-control study. Lett Appl Microbiol 2021; 74:498-504. [PMID: 34897759 DOI: 10.1111/lam.13630] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/05/2021] [Accepted: 12/06/2021] [Indexed: 01/04/2023]
Abstract
The pathogenesis of psoriasis, an immune-mediated chronic inflammatory skin disease, remains unclear. Studies have shown an association between psoriasis and intestinal inflammation; in this context, the influence of the gut microbiota on the immune response of psoriasis has become a focus of recent research. The present research evaluated the composition and diversity of the gut microbiota of 21 participants with psoriasis from a Brazilian referral dermatology service compared to 24 healthy controls. A stool sample was collected from each participant at the time of inclusion in the study, and the samples were analysed by sequencing the 16S rRNA gene. The recruitment of research participants involved matching between groups by sex, age, body mass index, comorbidities and smoking and the exclusion of several criteria that could potentially influence the gut microbiota and the interpretation of the data. There was an increase in the Dialister genus and Prevotella copri species in patients with psoriasis compared to the control group. A reduction in the Ruminococcus, Lachnospira and Blautia genera, as well as in the Akkermansia muciniphila species, was also verified in the psoriasis group compared to the control group. Furthermore, patients with psoriasis exhibited less gut microbiota diversity than controls.
Collapse
Affiliation(s)
- L Schade
- Department of Internal Medicine, Federal University of Paraná, Curitiba, Brazil
| | - D Mesa
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba-Paraná, Brazil
| | - A R Faria
- Department of Dermatology, Santa Casa de Misericórdia Hospital School of Medicine, Pontifical Catholic University of Paraná, Curitiba, Brazil
| | - J R Santamaria
- Department of Internal Medicine, Federal University of Paraná, Curitiba, Brazil
| | - C A Xavier
- Federal University of Paraná, Curitiba, Brazil
| | - D Ribeiro
- Federal University of Paraná, Curitiba, Brazil
| | - F N Hajar
- Federal University of Paraná, Curitiba, Brazil
| | - V F Azevedo
- Department of Internal Medicine, Federal University of Paraná, Curitiba, Brazil
| |
Collapse
|