1
|
Midya V, Nagdeo K, Lane JM, Torres-Olascoaga LA, Martínez GG, Horton MK, McRae N, Lopez I, Landero J, Gennings C, Téllez-Rojo MM, Wright RO, Arora M, Eggers S. Akkermansia muciniphila attenuates association between specific metal exposures during pregnancy and depressive symptoms in late childhood. iScience 2024; 27:111335. [PMID: 39640590 PMCID: PMC11617302 DOI: 10.1016/j.isci.2024.111335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/23/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Emerging research suggests that exposures to metals during pregnancy and consequent disruptions in gut microbiome (GM) are associated with depressive disorders in childhood. Akkermansia muciniphila, a GM bacteria, has been studied for its potential antidepressant effects. However, its role in influencing the association between prenatal metal exposures and depressive symptoms during childhood is unknown. Leveraging a well-characterized pediatric birth cohort and its microbiome substudy (n = 112), we investigated whether a certain subgroup of children at 9-11-year-of-age (characterized by a specific pattern of prenatal exposure to groups of metals or metal-clique) had worsened depressive symptoms and if the presence of A.muciniphila in GM modifies this association. A subgroup of children characterized by the prenatal metal-clique signature of zinc-chromium-cobalt had significantly increased depression scores; however, within that subgroup, children with A.muciniphila had much lower depression scores than those without A.muciniphila in the GM. Our analysis provides exploratory evidence hypothesizing A.muciniphila as an intervention attenuating the effect of prenatal metal-exposures-associated depressive disorders in late childhood.
Collapse
Affiliation(s)
- Vishal Midya
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiran Nagdeo
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jamil M. Lane
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Libni A. Torres-Olascoaga
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Mexico
| | - Gabriela Gil Martínez
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Mexico
| | - Megan K. Horton
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nia McRae
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Inessa Lopez
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julio Landero
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chris Gennings
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martha Maria Téllez-Rojo
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Mexico
| | - Robert O. Wright
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manish Arora
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Linus Biotechnology, Inc., North Brunswick Township, NJ, USA
| | - Shoshannah Eggers
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Epidemiology, University of Iowa College of Public Health, Iowa City, IA, USA
| |
Collapse
|
2
|
Ribes-Martínez L, Muñoz-Egea MC, Yuste J, Esteban J, García-Quintanilla M. Bacteriophage Therapy as a Promising Alternative for Antibiotic-Resistant Enterococcus faecium: Advances and Challenges. Antibiotics (Basel) 2024; 13:1120. [PMID: 39766510 PMCID: PMC11672805 DOI: 10.3390/antibiotics13121120] [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: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
Enterococcus faecium is a Gram-positive bacterium increasingly identified as a critical nosocomial pathogen that poses significant treatment challenges due to its resistance to multiple antibiotics, particularly vancomycin-resistant E. faecium (VRE) strains. The urgent need for alternative therapeutic strategies has renewed interest in bacteriophage (phage) therapy, given phages specificity and bactericidal potential. This review explores the advancements in phage therapy against antibiotic-resistant E. faecium, including phage morphological diversity, genomic characteristics, and infection mechanisms. The efficacy of phage therapy in in vitro, ex vivo, and in vivo models and the compassionate use in clinical settings are evaluated, highlighting the promising outcomes of phage-antibiotic synergies and biofilm disruption. Key challenges and future research directions are discussed, with a focus on improving therapeutic efficacy and overcoming bacterial resistance. This review emphasizes the potential of phage therapy as a viable solution for managing multidrug-resistant E. faecium infections and underscores the importance of future investigations to enhance clinical applications.
Collapse
Affiliation(s)
- Laura Ribes-Martínez
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (L.R.-M.); (M.-C.M.-E.); (J.E.)
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
- MePRAM, Proyecto de Medicina de Precisión Contra las Resistencias Antimicrobianas, CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Maria-Carmen Muñoz-Egea
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (L.R.-M.); (M.-C.M.-E.); (J.E.)
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| | - Jose Yuste
- MePRAM, Proyecto de Medicina de Precisión Contra las Resistencias Antimicrobianas, CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Spanish Pneumococcal Reference Laboratory, National Center for Microbiology, Instituto de Salud Carlos III, 28029 Madrid, Spain
- CIBERES-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jaime Esteban
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (L.R.-M.); (M.-C.M.-E.); (J.E.)
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
- MePRAM, Proyecto de Medicina de Precisión Contra las Resistencias Antimicrobianas, CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Meritxell García-Quintanilla
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (L.R.-M.); (M.-C.M.-E.); (J.E.)
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
- MePRAM, Proyecto de Medicina de Precisión Contra las Resistencias Antimicrobianas, CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| |
Collapse
|
3
|
Emanowicz P, Średnicka P, Wójcicki M, Roszko M, Juszczuk-Kubiak E. Mitigating Dietary Bisphenol Exposure Through the Gut Microbiota: The Role of Next-Generation Probiotics in Bacterial Detoxification. Nutrients 2024; 16:3757. [PMID: 39519589 PMCID: PMC11547510 DOI: 10.3390/nu16213757] [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: 09/03/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Bisphenols, such as bisphenol A and its analogs, which include bisphenol S, bisphenol F, bisphenol AF, and tetramethyl bisphenol F, are chemical contaminants commonly found in food that raise serious health concerns. These xenobiotics can potentially have harmful effects on human health. The gut microbiota plays a crucial role in metabolizing and neutralizing these substances, which is essential for their detoxification and elimination. Probiotic supplementation has been studied for its ability to modulate the gut microbiota's composition and function, enhancing detoxification processes. Next-Generation Probiotics (NGPs) may exhibit better properties than traditional strains and are designed for targeted action on specific conditions, such as obesity. By modulating inflammatory responses and reducing the secretion of pro-inflammatory cytokines, they can significantly improve host health. Research on NGPs' ability to neutralize obesogenic bisphenols remains limited, but their potential makes this a promising area for future exploration. This review aims to understand the mechanisms of the chemical transformation of bisphenol through its interactions with the gut microbiota and the role of probiotics, particularly NGPs, in these processes. Understanding the interplay between bisphenols, gut microbiota, and NGPs may pave the way for strategies to counteract the negative health effects associated with daily and chronic exposure to bisphenols, which is crucial for food safety and consumer health protection.
Collapse
Affiliation(s)
- Paulina Emanowicz
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology–State Research Institute, Rakowiecka 36 Street, 02-532 Warsaw, Poland; (P.Ś.); (M.W.); (E.J.-K.)
| | - Paulina Średnicka
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology–State Research Institute, Rakowiecka 36 Street, 02-532 Warsaw, Poland; (P.Ś.); (M.W.); (E.J.-K.)
| | - Michał Wójcicki
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology–State Research Institute, Rakowiecka 36 Street, 02-532 Warsaw, Poland; (P.Ś.); (M.W.); (E.J.-K.)
| | - Marek Roszko
- Department of Food Safety and Chemical Analysis, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology–State Research Institute, Rakowiecka 36 Street, 02-532 Warsaw, Poland;
| | - Edyta Juszczuk-Kubiak
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology–State Research Institute, Rakowiecka 36 Street, 02-532 Warsaw, Poland; (P.Ś.); (M.W.); (E.J.-K.)
| |
Collapse
|
4
|
Midya V, Nagdeo K, Lane J, Torres-Olascoaga L, Martínez G, Horton M, Gennings C, Téllez-Rojo M, Wright R, Arora M, Eggers S. Akkermansia muciniphila modifies the association between metal exposure during pregnancy and depressive symptoms in late childhood. RESEARCH SQUARE 2024:rs.3.rs-3922286. [PMID: 38410473 PMCID: PMC10896378 DOI: 10.21203/rs.3.rs-3922286/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Emerging research suggests that exposures to metals during pregnancy and gut microbiome (GM) disruptions are associated with depressive disorders in childhood. Akkermansia muciniphila, a GM bacteria, has been studied for its potential antidepressant effects. However, its role in the influence of prenatal metal exposures on depressive symptoms during childhood is unknown. Leveraging a well-characterized pediatric longitudinal birth cohort and its microbiome substudy (n=112) and using a state-of-the-art machine-learning model, we investigated whether the presence of A.muciniphila in GM of 9-11-year-olds modifies the associations between exposure to a specific group of metals (or metal-clique) during pregnancy and concurrent childhood depressive symptoms. Among children with no A.muciniphila, a metal-clique of Zinc-Chromium-Cobalt was strongly associated with increased depression score (P<0.0001), whereas, for children with A.muciniphila, this same metal-clique was weakly associated with decreased depression score(P<0.4). Our analysis provides the first exploratory evidence hypothesizing A. muciniphila as a probiotic intervention attenuating the effect of prenatal metal-exposures-associated depressive disorders in late childhood.
Collapse
Affiliation(s)
| | | | | | | | - Gabriela Martínez
- Center for Research on Nutrition and Health, National Institute of Public Health
| | | | | | - Martha Téllez-Rojo
- Center for Research on Nutrition and Health, National Institute of Public Health
| | | | | | | |
Collapse
|
5
|
Kumaria A, Kamaludin A, Ashkan K. Nutritional adjuncts in the management of gliomas: Food for thought. Clin Neurol Neurosurg 2024; 236:108102. [PMID: 38157656 DOI: 10.1016/j.clineuro.2023.108102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
|
6
|
Sparfel L, Ratodiarivony S, Boutet-Robinet E, Ellero-Simatos S, Jolivet-Gougeon A. Akkermansia muciniphila and Alcohol-Related Liver Diseases. A Systematic Review. Mol Nutr Food Res 2024; 68:e2300510. [PMID: 38059838 DOI: 10.1002/mnfr.202300510] [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: 07/18/2023] [Revised: 10/03/2023] [Indexed: 12/08/2023]
Abstract
SCOPE Akkermansia muciniphila (A. muciniphila) are Gram negative commensal bacteria, degrading mucin in the intestinal mucosa, modulating intestinal permeability and inflammation in the digestive tract, liver, and blood. Some components can promote the relative abundance of A. muciniphila in the gut microbiota, but lower levels of A. muciniphila are more commonly found in people with obesity, diabetes, metabolic syndromes, or inflammatory digestive diseases. Over-intake of ethanol can also induce a decrease of A. muciniphila, associated with dysregulation of microbial metabolite production, impaired intestinal permeability, induction of chronic inflammation, and production of cytokines. METHODS AND RESULTS Using a PRISMA search strategy, a review is performed on the bacteriological characteristics of A. muciniphila, the factors capable of modulating its relative abundance in the digestive tract and its probiotic use in alcohol-related liver diseases (alcoholic hepatitis, cirrhosis, hepatocellular carcinoma, hepatic transplantation, partial hepatectomy). CONCLUSION Several studies have shown that supplementation with A. muciniphila can improve ethanol-related hepatic pathologies, and highlight the interest in using this bacterial species as a probiotic.
Collapse
Affiliation(s)
- Lydie Sparfel
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, F-35000, France
| | - Sandy Ratodiarivony
- Univ Rennes, Bacterial Regulatory RNAs and Medicine (BRM), UMR_S 1230, Rennes, F-35000, France
| | - Elisa Boutet-Robinet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300, Toulouse, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300, Toulouse, France
| | - Anne Jolivet-Gougeon
- Univ Rennes, Bacterial Regulatory RNAs and Medicine (BRM), UMR_S 1230, Rennes, F-35000, France
- Teaching Hospital, CHU Rennes, 2 rue Henri Le Guilloux 35033, Rennes, F-35000, France
- INSERM, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer), U1241, INSERM 1241, Rennes, F-35000, France
| |
Collapse
|
7
|
Pan LL, Ren ZN, Yang J, Li BB, Huang YW, Song DX, Li X, Xu JJ, Bhatia M, Zou DW, Zhou CH, Sun J. Gut microbiota controls the development of chronic pancreatitis: A critical role of short-chain fatty acids-producing Gram-positive bacteria. Acta Pharm Sin B 2023; 13:4202-4216. [PMID: 37799394 PMCID: PMC10547962 DOI: 10.1016/j.apsb.2023.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/21/2023] [Accepted: 06/13/2023] [Indexed: 10/07/2023] Open
Abstract
Chronic pancreatitis (CP) is a progressive and irreversible fibroinflammatory disorder, accompanied by pancreatic exocrine insufficiency and dysregulated gut microbiota. Recently, accumulating evidence has supported a correlation between gut dysbiosis and CP development. However, whether gut microbiota dysbiosis contributes to CP pathogenesis remains unclear. Herein, an experimental CP was induced by repeated high-dose caerulein injections. The broad-spectrum antibiotics (ABX) and ABX targeting Gram-positive (G+) or Gram-negative bacteria (G-) were applied to explore the specific roles of these bacteria. Gut dysbiosis was observed in both mice and in CP patients, which was accompanied by a sharply reduced abundance for short-chain fatty acids (SCFAs)-producers, especially G+ bacteria. Broad-spectrum ABX exacerbated the severity of CP, as evidenced by aggravated pancreatic fibrosis and gut dysbiosis, especially the depletion of SCFAs-producing G+ bacteria. Additionally, depletion of SCFAs-producing G+ bacteria rather than G- bacteria intensified CP progression independent of TLR4, which was attenuated by supplementation with exogenous SCFAs. Finally, SCFAs modulated pancreatic fibrosis through inhibition of macrophage infiltration and M2 phenotype switching. The study supports a critical role for SCFAs-producing G+ bacteria in CP. Therefore, modulation of dietary-derived SCFAs or G+ SCFAs-producing bacteria may be considered a novel interventive approach for the management of CP.
Collapse
Affiliation(s)
- Li-Long Pan
- Wuxi Medical School, Jiangnan University, Wuxi 214122, China
| | - Zheng-Nan Ren
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Jun Yang
- Department of General Surgery, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Bin-Bin Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Yi-Wen Huang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Dong-Xiao Song
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Xuan Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Jia-Jia Xu
- Department of General Medicine, Beicai Community Health Service Center of Pudong New District, Shanghai 214001, China
| | - Madhav Bhatia
- Department of Pathology, University of Otago, Christchurch 8140, New Zealand
| | - Duo-Wu Zou
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chun-Hua Zhou
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jia Sun
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
8
|
Yamamoto-Hanada K, Sato M, Toyokuni K, Irahara M, Hiraide-Kotaki E, Harima-Mizusawa N, Morita H, Matsumoto K, Ohya Y. Combination of heat-killed Lactiplantibacillus plantarum YIT 0132 (LP0132) and oral immunotherapy in cow's milk allergy: a randomised controlled trial. Benef Microbes 2023; 14:17-30. [PMID: 36815492 DOI: 10.3920/bm2022.0064] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Safer and more effective cow milk (CM)-oral immunotherapy that does not induce allergic reactions has not yet been standardised. We sought to explore the efficacy and feasibility of a combination of heat-killed Lactiplantibacillus plantarum YIT 0132 (LP0132) and oral immunotherapy for treating IgE-mediated cow milk allergy (CMA). We conducted a 24-week, double-blind, randomised (1:1), two-arm, parallel-group, placebo-controlled, phase 2 trial of LP0132 intervention for treating IgE-mediated CMA in children aged 1-18 years (n=60) from January 29, 2018 to July 12, 2019 in Tokyo, Japan. Participants were randomly assigned to the LP0132 group receiving citrus juice fermented with LP0132 or to the control group receiving citrus juice without. Both groups received low-dose slow oral immunotherapy with CM. The primary outcome was improved tolerance to CM, proven by the CM challenge test at 24 weeks. Secondary outcomes were changes in serum biomarkers of serum-specific β-lactoglobulin-IgE (sIgE) and β-lactoglobulin-IgG4 (sIgG4). Exploratory outcomes included changes in serum cytokine levels and gut microbiota composition. A total of 61 participants were included. Finally, 31 children were assigned to the LP0132 group and 30 to the control group, respectively. After the intervention, 41.4 and 37.9% of the participants in the LP0132 and control groups, respectively, showed improved tolerance to CM. In serum biomarkers after the intervention, the sIgG4 level was significantly higher, and interleukin (IL)-5 and IL-9 were significantly lower, in the LP0132 group than in the control group. In the gut microbiome, the α-diversity and Lachnospiraceae increased significantly in the LP0132 group, and Lachnospiraceae after the intervention was significantly higher in the LP0132 group than in the control group. In conclusion, low-dose oral immunotherapy with modulating gut microbiota might be a safer and more effective approach for treating cow's milk allergy.
Collapse
Affiliation(s)
- K Yamamoto-Hanada
- Allergy Center, National Center for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| | - M Sato
- Allergy Center, National Center for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| | - K Toyokuni
- Allergy Center, National Center for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| | - M Irahara
- Allergy Center, National Center for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| | - E Hiraide-Kotaki
- Yakult Central Institute for Microbiological Research, 5-11 Izumi, Kunitachi, Tokyo 186-8650, Japan
| | - N Harima-Mizusawa
- Yakult Central Institute for Microbiological Research, 5-11 Izumi, Kunitachi, Tokyo 186-8650, Japan
| | - H Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| | - K Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| | - Y Ohya
- Allergy Center, National Center for Child Health and Development, 2-10-1 Okura, 1578535 Tokyo, Japan
| |
Collapse
|
9
|
Rodpai R, Sanpool O, Janwan P, Boonroumkaew P, Sadaow L, Thanchomnang T, Intapan PM, Maleewong W. Gut microbiota diversity in human strongyloidiasis differs little in two different regions in endemic areas of Thailand. PLoS One 2022; 17:e0279766. [PMID: 36584127 PMCID: PMC9803247 DOI: 10.1371/journal.pone.0279766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
Human gastrointestinal helminthic infections have a direct and/or indirect effect on the composition of the host gut microbial flora. Here, we investigated the effect of infection with a soil-transmitted intestinal nematode, Strongyloides stercoralis, on the gut microbiota of the human host. We also investigated whether composition of the microbiota in infected persons might vary across endemic regions. Fecal samples were obtained from volunteers from two areas endemic for strongyloidiasis, Khon Kaen Province in northeastern Thailand and Nakhon Si Thammarat Province in southern Thailand. Samples from Khon Kaen were from infected (SsNE) and uninfected (NegNE) individuals. Similarly, samples from the latter province were from infected (SsST) and uninfected (NegST) individuals. DNA sequences of the V3-V4 regions of the bacterial 16S rRNA gene were obtained from the fecal samples. No statistical difference in alpha diversity between groups in terms of richness or diversity were found. Statistical difference in beta diversity was observed only between NegNE and NegST. Some significant differences in species abundance were noted between geographical isolates. The SsNE group had a higher abundance of Tetragenococcus holophilus than did the SsST group, whereas Bradyrhizobium sp. was less abundant in the SsNE than the SsST group. For the uninfected groups, the NegNE had a higher abundance of T. holophilus than the NegST group. Our data showed that S. stercoralis infection leads to only minor alterations in the relative abundance of individual bacterial species in the human gut: no detectable effect was observed on community structure and diversity.
Collapse
Affiliation(s)
- Rutchanee Rodpai
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Oranuch Sanpool
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Penchom Janwan
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand
| | - Patcharaporn Boonroumkaew
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Lakkhana Sadaow
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Tongjit Thanchomnang
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Faculty of Medicine, Mahasarakham University, Mahasarakham, Thailand
| | - Pewpan M. Intapan
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Wanchai Maleewong
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
- * E-mail:
| |
Collapse
|
10
|
Sheng S, Fu Y, Pan N, Zhang H, Xiu L, Liang Y, Liu Y, Liu B, Ma C, Du R, Wang X. Novel exopolysaccharide derived from probiotic Lactobacillus pantheris TCP102 strain with immune-enhancing and anticancer activities. Front Microbiol 2022; 13:1015270. [PMID: 36225355 PMCID: PMC9549278 DOI: 10.3389/fmicb.2022.1015270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Probiotics are gaining attention due to their functions of regulating the intestinal barrier and promoting human health. The production of exopolysaccharide (EPS) is one of the important factors for probiotics to exert beneficial properties. This study aimed to screen exopolysaccharides-producing lactic acid bacteria (LAB) and evaluate the probiotic potential. we obtained three exopolysaccharide fractions (EPS1, EPS2, and EPS3) from Lactobacillus pantheris TCP102 and purified by a combination of ion-exchange chromatography and gel permeation chromatography. The structures of the fractions were characterized by FT-IR, UV, HPLC, and scanning electron microscopy (SEM) analysis. The Mw of EPS1, EPS2, and EPS3 were approximately 20.3, 23.0, and 19.3 kDa, and were mainly composed of galactose, glucose, and mannose, with approximate molar ratios of 2.86:1:1.48, 1.26:1:1, 1.58:1.80:1, respectively. Furthermore, SEM analysis demonstrated that the three polysaccharide fractions differ in microstructure and surface morphology. Additionally, preliminary results for immune-enhancing and anticancer activities reveal that these EPSs significantly induced the production of nitric oxide (NO), TNF-α, and IL-6 in Ana-1 cells and peritoneal macrophage cells. Meanwhile, the EPSs also significantly suppressed the proliferation of HCT-116, BCG-803, and particularly A-2780 cells. The results suggest that the three novel EPSs isolated from Lactobacillus pantheris TCP102 can be regarded as potential application value in functional food and natural antitumor drugs.
Collapse
Affiliation(s)
- Shouxin Sheng
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yubing Fu
- School of Life Sciences, Faculty of Medicine and Life Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, China
| | - Na Pan
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Haochi Zhang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Lei Xiu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yanchen Liang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yang Liu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Bohui Liu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Cheng Ma
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Ruiping Du
- Animal Nutrition Institute, Agriculture and Animal Husbandry Academy of Inner Mongolia, Hohhot, China
| | - Xiao Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| |
Collapse
|
11
|
Tian B, Yao JH, Lin X, Lv WQ, Jiang LD, Wang ZQ, Shen J, Xiao HM, Xu H, Xu LL, Cheng X, Shen H, Qiu C, Luo Z, Zhao LJ, Yan Q, Deng HW, Zhang LS. Metagenomic study of the gut microbiota associated with cow milk consumption in Chinese peri-/postmenopausal women. Front Microbiol 2022; 13:957885. [PMID: 36051762 PMCID: PMC9425034 DOI: 10.3389/fmicb.2022.957885] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
Cow milk consumption (CMC) and alterations of gut bacterial composition are proposed to be closely related to human health and disease. Our research aims to investigate the changes in human gut microbial composition in Chinese peri-/postmenopausal women with different CMC habits. A total of 517 subjects were recruited and questionnaires about their CMC status were collected; 394 subjects were included in the final analyses. Fecal samples were used for studying gut bacterial composition. All the subjects were divided into a control group (n = 248) and a CMC group (n = 146) according to their CMC status. Non-parametric tests and LEfSe at different taxonomic levels were used to reveal differentially abundant taxa and functional categories across different CMC groups. Relative abundance (RA) of one phylum (p_Actinobacteria), three genera (g_Bifidobacterium, g_Anaerostipes, and g_Bacteroides), and 28 species diversified significantly across groups. Specifically, taxa g_Anaerostipes (p < 0.01), g_Bacteroides (p < 0.05), s_Anaerostipes_hadrus (p < 0.01), and s_Bifidobacterium_pseudocatenulatum (p < 0.01) were positively correlated with CMC levels, but p_Actinobacteria (p < 0.01) and g_Bifidobacterium (p < 0.01) were negatively associated with CMC levels. KEGG module analysis revealed 48 gut microbiome functional modules significantly (p < 0.05) associated with CMC, including Vibrio cholerae pathogenicity signature, cholera toxins (p = 9.52e-04), and cephamycin C biosynthesis module (p = 0.0057), among others. In conclusion, CMC was associated with changes in gut microbiome patterns including beta diversity and richness of some gut microbiota. The alterations of certain bacteria including g_Anaerostipes and s_Bifidobacterium_pseudocatenulatum in the CMC group should be important for human health. This study further supports the biological value of habitual cow milk consumption.
Collapse
Affiliation(s)
- Bo Tian
- School of Physical Science and Engineering, College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Jia-Heng Yao
- School of Physical Science and Engineering, College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Xu Lin
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Wan-Qiang Lv
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, China
| | - Lin-Dong Jiang
- Tulane Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Zhuo-Qi Wang
- School of Physical Science and Engineering, College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Jie Shen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China,Shunde Hospital of Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
| | - Hong-Mei Xiao
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, China
| | - Hanli Xu
- School of Physical Science and Engineering, College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Lu-Lu Xu
- School of Physical Science and Engineering, College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Xiyu Cheng
- School of Physical Science and Engineering, College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Hui Shen
- Tulane Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Chuan Qiu
- Tulane Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Zhe Luo
- Tulane Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Lan-Juan Zhao
- Tulane Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Qiong Yan
- School of Physical Science and Engineering, College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Hong-Wen Deng
- Tulane Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States,*Correspondence: Li-Shu Zhang,
| | - Li-Shu Zhang
- School of Physical Science and Engineering, College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China,Hong-Wen Deng,
| |
Collapse
|
12
|
Wang S, Huang G, Wang JX, Tian L, Zuo XL, Li YQ, Yu YB. Altered Gut Microbiota in Patients With Peutz–Jeghers Syndrome. Front Microbiol 2022; 13:881508. [PMID: 35910641 PMCID: PMC9326469 DOI: 10.3389/fmicb.2022.881508] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/06/2022] [Indexed: 11/14/2022] Open
Abstract
Background Peutz–Jeghers syndrome (PJS) is a rare genetic disorder characterized by the development of pigmented spots and gastrointestinal polyps and increased susceptibility to cancers. It remains unknown whether gut microbiota dysbiosis is linked to PJS. Aim This study aimed to assess the structure and composition of the gut microbiota, including both bacteria and fungi, in patients with PJS and investigate the relationship between gut microbiota dysbiosis and PJS pathogenesis. Methods The bacterial and fungal composition of the fecal microbiota was analyzed in 23 patients with PJS (cases), 17 first-degree asymptomatic relatives (ARs), and 24 healthy controls (HCs) using 16S (MiSeq) and ITS2 (pyrosequencing) sequencing for bacteria and fungi, respectively. Differential analyses of the intestinal flora were performed from the phylum to species level. Results Alpha-diversity distributions of bacteria and fungi indicated that the abundance of both taxa differed between PJS cases and controls. However, while the diversity and composition of fecal bacteria in PJS cases were significantly different from those in ARs and HCs, fungal flora was more stable. High-throughput sequencing confirmed the special characteristics and biodiversity of the fecal bacterial and fungal microflora in patients with PJS. They had lower bacterial biodiversity than controls, with a higher frequency of the Proteobacteria phylum, Enterobacteriaceae family, and Escherichia-Shigella genus, and a lower frequency of the Firmicutes phylum and the Lachnospiraceae and Ruminococcaceae families. Of fungi, Candida was significantly higher in PJS cases than in controls. Conclusion The findings reported here confirm gut microbiota dysbiosis in patients with PJS. This is the first report on the bacterial and fungal microbiota profile of subjects with PJS, which may be meaningful to provide a structural basis for further research on intestinal microecology in PJS.
Collapse
Affiliation(s)
- Sui Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Gang Huang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Jue-Xin Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Tian
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiu-Li Zuo
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Yan-Qing Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Yan-Bo Yu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Yan-Bo Yu
| |
Collapse
|
13
|
Beneficial metabolic effects of probiotic supplementation in dams and offspring following hypercaloric diet during pregnancy. NUTR CLIN METAB 2022. [DOI: 10.1016/j.nupar.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
14
|
Kamil RZ, Murdiati A, Juffrie M, Rahayu ES. Gut Microbiota Modulation of Moderate Undernutrition in Infants through Gummy Lactobacillus plantarum Dad-13 Consumption: A Randomized Double-Blind Controlled Trial. Nutrients 2022; 14:1049. [PMID: 35268024 PMCID: PMC8912314 DOI: 10.3390/nu14051049] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/10/2022] [Accepted: 02/25/2022] [Indexed: 01/03/2023] Open
Abstract
Undernutrition is associated with gut microbiota unbalance, and probiotics are believed to restore it and improve gut integrity. A randomized double-blind controlled trial was conducted to evaluate the efficacy of gummy L. plantarum Dad-13 (108-9 CFU/3 g) to prevent the progression of severe undernutrition. Two groups of moderate undernutrition infants were involved in this study, namely the placebo (n = 15) and probiotics (n = 15) groups, and were required to consume the product for 50 days. 16S rRNA sequencing and qPCR were used for gut microbiota analysis, and gas chromatography was used to analyze Short-Chain Fatty Acid (SCFA). The daily food intake of both groups was recorded using food records. Our results revealed that the probiotic group had better improvements regarding the anthropometry and nutritional status. In addition, L. plantarum Dad-13 modulated the butyric acid-producing bacteria to increase and inhibit the growth of Enterobacteriaceae. This gut modulation was associated with the increment in SCFA, especially total SCFA, propionic, and butyric acid. The number of L. plantarum was increased after the probiotic intervention. However, L. plantarum Dad-13 was not able to change the alpha and beta diversity. Therefore, L. plantarum Dad-13 has been proven to promote the growth of beneficial bacteria.
Collapse
Affiliation(s)
- Rafli Zulfa Kamil
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Jl. Flora No 1 Bulaksumur, Yogyakarta 55281, Indonesia; (R.Z.K.); (A.M.)
- Centre for Food and Nutrition Studies, Universitas Gadjah Mada, Jl. Teknika Utara Barek, Yogyakarta 55281, Indonesia
- Centre of Excellence for Probiotics, Universitas Gadjah Mada, Jl. Teknika Utara Barek, Yogyakarta 55281, Indonesia
- Department of Food Technology, Faculty of Animal and Agricultural Sciences, Universitas Diponegoro, Jl. Prof. Soedarto, Tembalang, Semarang 50275, Indonesia
| | - Agnes Murdiati
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Jl. Flora No 1 Bulaksumur, Yogyakarta 55281, Indonesia; (R.Z.K.); (A.M.)
| | - Mohammad Juffrie
- Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Jl. Farmako, Senolowo, Sekip Utara, Yogyakarta 55281, Indonesia;
| | - Endang Sutriswati Rahayu
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Jl. Flora No 1 Bulaksumur, Yogyakarta 55281, Indonesia; (R.Z.K.); (A.M.)
- Centre for Food and Nutrition Studies, Universitas Gadjah Mada, Jl. Teknika Utara Barek, Yogyakarta 55281, Indonesia
- Centre of Excellence for Probiotics, Universitas Gadjah Mada, Jl. Teknika Utara Barek, Yogyakarta 55281, Indonesia
| |
Collapse
|
15
|
Wang Y, Dong J, Wang J, Chi W, Zhou W, Tian Q, Hong Y, Zhou X, Ye H, Tian X, Hu R, Wong A. Assessing the drug resistance profiles of oral probiotic lozenges. J Oral Microbiol 2022; 14:2019992. [PMID: 35024089 PMCID: PMC8745366 DOI: 10.1080/20002297.2021.2019992] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Probiotic lozenges have been developed to harvest the benefits of probiotics for oral health, but their long-term consumption may encourage the transfer of resistance genes from probiotics to commensals, and eventually to disease-causing bacteria. Aim To screen commercial probiotic lozenges for resistance to antibiotics, characterize the resistance determinants, and examine their transferability in vitro. Results Probiotics of all lozenges were resistant to glycopeptide, sulfonamide, and penicillin antibiotics, while some were resistant to aminoglycosides and cephalosporins. High minimum inhibitory concentrations (MICs) were detected for streptomycin (>128 µg/mL) and chloramphenicol (> 512 µg/mL) for all probiotics but only one was resistant to piperacillin (MIC = 32 µg/mL). PCR analysis detected erythromycin (erm(T), ermB or mefA) and fluoroquinolone (parC or gyr(A)) resistance genes in some lozenges although there were no resistant phenotypes. The dfrD, cat-TC, vatE, aadE, vanX, and aph(3")-III or ant(2")-I genes conferring resistance to trimethoprim, chloramphenicol, quinupristin/dalfopristin, vancomycin, and streptomycin, respectively, were detected in resistant probiotics. The rifampicin resistance gene rpoB was also present. We found no conjugal transfer of streptomycin resistance genes in our co-incubation experiments. Conclusion Our study represents the first antibiotic resistance profiling of probiotics from oral lozenges, thus highlighting the health risk especially in the prevailing threat of drug resistance globally.
Collapse
Affiliation(s)
- Yi Wang
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, University Town, Wenzhou, Zhejiang Province, China
| | - Jingya Dong
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, University Town, Wenzhou, Zhejiang Province, China
| | - Junyi Wang
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Wei Chi
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Wei Zhou
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Qiwen Tian
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Yue Hong
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, University Town, Wenzhou, Zhejiang Province, China
| | - Xuan Zhou
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Hailv Ye
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Xuechen Tian
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou, Zhejiang Province, China.,Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou, Zhejiang Province, China
| | - Rongdang Hu
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, University Town, Wenzhou, Zhejiang Province, China
| | - Aloysius Wong
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China.,Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou, Zhejiang Province, China.,Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou, Zhejiang Province, China
| |
Collapse
|
16
|
Li Y, Yi J, Zeng Q, Liu Y, Yang B, Liu B, Li Y, Mehmood K, Hussain R, Tang Z, Zhang H, Li Y. Zearalenone exposure mediated hepatotoxicity via mitochondrial apoptotic and autophagy pathways: Associated with gut microbiome and metabolites. Toxicology 2021; 462:152957. [PMID: 34537261 DOI: 10.1016/j.tox.2021.152957] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/07/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022]
Abstract
Zearalenone (ZEN), a mycotoxin is frequently detected in different food products and has been widely studied for its toxicity. However, the underlying mechanisms of hepatotoxic effects, relationship between gut microbiome and liver metabolite mediated hepatotoxicity mechanisms induced by ZEN are still not clear. Here, we reported that the different microscopic changes like swelling of hepatocyte, disorganization of hepatocytes and extensive vacuolar degeneration were observed, and the mitochondrial functions decreased in exposed mice. Results exhibited up-regulation in expression of signals of apoptosis and autophagy in liver of treated mice via mitochondrial apoptotic and autophagy pathway (Beclin1/p62). The diversity of gut microbiome decreased and the values of various microbiome altered in treated mice, including 5 phyla (Chloroflexi, Sva0485, Methylomirabilota, MBNT15 and Kryptonia) and genera (Frankia, Lactococcus, Anaerolinea, Halomonas and Sh765B-TzT-35) significantly changed. Liver metabolism showed that the concentrations of 91 metabolite including lipids and lipid like molecules were significantly changed. The values of phosphatidylcholine, 2-Lysophosphatidylcholine and phosphatidate concentrations suggestive of abnormal glycerophosphate metabolism pathway were significantly increased in mice due to exposure to ZEN. In conclusion, the findings suggest that the disorders in gut microbiome and liver metabolites due to exposure to ZEN in mice may affect the liver.
Collapse
Affiliation(s)
- Yuanliang Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jiangnan Yi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Qiwen Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yingwei Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Bijing Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Bingxian Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yangwei Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Khalid Mehmood
- Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Riaz Hussain
- Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
17
|
Alteration of the gut fecal microbiome in children living with HIV on antiretroviral therapy in Yaounde, Cameroon. Sci Rep 2021; 11:7666. [PMID: 33828220 PMCID: PMC8027858 DOI: 10.1038/s41598-021-87368-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 03/15/2021] [Indexed: 02/08/2023] Open
Abstract
Multiple factors, such as immune disruption, prophylactic co-trimoxazole, and antiretroviral therapy, may influence the structure and function of the gut microbiome of children infected with HIV from birth. In order to understand whether HIV infection altered gut microbiome and to relate changes in microbiome structure and function to immune status, virological response and pediatric ART regimens, we characterized the gut microbiome of 87 HIV-infected and 82 non-exposed HIV-negative children from Yaounde, a cosmopolitan city in Cameroon. We found that children living with HIV had significantly lower alpha diversity in their gut microbiome and altered beta diversity that may not be related to CD4+ T cell count or viral load. There was an increased level of Akkermansia and Faecalibacterium genera and decreased level of Escherichia and other Gamma proteobacteria in children infected with HIV, among other differences. We noted an effect of ethnicity/geography on observed gut microbiome composition and that children on ritonavir-boosted protease inhibitor (PI/r)-based ART had gut microbiome composition that diverged more from HIV-negative controls compared to those on non-nucleoside reverse-transcriptase inhibitors-based ART. Further studies investigating the role of this altered gut microbiome in increased disease susceptibility are warranted for individuals who acquired HIV via mother-to-child transmission.
Collapse
|
18
|
Collinet A, Grimm P, Julliand S, Julliand V. Multidimensional Approach for Investigating the Effects of an Antibiotic-Probiotic Combination on the Equine Hindgut Ecosystem and Microbial Fibrolysis. Front Microbiol 2021; 12:646294. [PMID: 33841371 PMCID: PMC8027512 DOI: 10.3389/fmicb.2021.646294] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/16/2021] [Indexed: 12/11/2022] Open
Abstract
The equine hindgut ecosystem is specialized in dietary fibers' fermentation to provide horses' energy and contribute to its health. Nevertheless, antibiotics are known to disrupt the hindgut microbiota, affecting the fibrolytic activity of bacteria and the intestinal immune balance, leading to diseases. This in vivo study used a general and comprehensive approach for characterizing the hindgut ecosystem of 9 healthy horses over 28 days in response to a 5-day challenge with oral trimethoprim-sulfadiazine (TMS), with a special emphasis on microbial fibrolytic activity and the host immune response. Horses were supplemented with two doses of Lactobacillus acidophilus, Ligilactobacillus salivarius (formerly L. salivarius), and Bifidobacterium lactis blend or a placebo in a 3 × 3 Latin square design. Changes in fecal microbiota were investigated using 16S rRNA sequencing. Clostridioides difficile was quantified in feces using quantitative polymerase chain reaction. Anaerobic microbiological culture was used to enumerate functional bacterial groups (cellulolytic, amylolytic, and lactic acid-utilizing). The environmental dimensions were assessed by measuring the concentrations of volatile fatty acids (VFAs) and lactic acid using biochemical methods, and changes in pH and dry matter weight. Systemic and local inflammation was evaluated by determination of cytokine and immunoglobulin (Ig)A concentrations in the serum and secretory IgA (SIgA) concentrations in the feces using immuno-enzymatic methods. Oral TMS treatment strongly altered the whole hindgut ecosystem by 2 days after the first administration. Bacterial diversity decreased in proportion to the relative abundance of fibrolytic genera, which coincided with the decrease in the concentration of cellulolytic bacteria. At the same time, the composition of microbiota members was reorganized in terms of relative abundances, probably to support the alteration in fibrolysis. C. difficile DNA was not found in these horses, but the relative abundances of several potential pathobiont genera increased. 2 days after the first TMS administration, fecal concentrations of VFAs and SIgA increased in parallel with fecal water content, suggesting an alteration of the integrity of the hindgut mucosa. Recovery in bacterial composition, functions, and immune biomarkers took 2-9 days after the end of TMS administration. Supplementation with this bacterial blend did not limit bacterial alteration but might have interesting mucosal immunomodulatory effects.
Collapse
Affiliation(s)
- Axelle Collinet
- Lab To Field, Dijon, France.,Univ. Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, Dijon, France
| | | | | | - Véronique Julliand
- Univ. Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, Dijon, France
| |
Collapse
|
19
|
Kaur J, Singh BP, Chaudhary V, Elshaghabee FMF, Singh J, Singh A, Rokana N, Panwar H. Probiotics as Live Bio-therapeutics: Prospects and Perspectives. MICROORGANISMS FOR SUSTAINABILITY 2021:83-120. [DOI: 10.1007/978-981-15-6795-7_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
20
|
Zommiti M, Feuilloley MGJ, Connil N. Update of Probiotics in Human World: A Nonstop Source of Benefactions till the End of Time. Microorganisms 2020; 8:E1907. [PMID: 33266303 PMCID: PMC7760123 DOI: 10.3390/microorganisms8121907] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023] Open
Abstract
Lactic acid bacteria (LAB) are known for their biotechnological potential. Moreover, LAB are distinguished by amazing criteria: Adjusting the intestinal environment, inhibiting pathogenic microbes in the gastrointestinal tract, ability to reduce pathogen adhesion activity, improving the balance of the microbiota inside the intestine, capabilities of regulating intestinal mucosal immunity, and maintaining intestinal barrier function. The escalating number of research and studies about beneficial microorganisms and their impact on promoting health has attracted a big interest in the last decades. Since antiquity, various based fermented products of different kinds have been utilized as potential probiotic products. Nevertheless, the current upsurge in consumers' interest in bioalternatives has opened new horizons for the probiotic field in terms of research and development. The present review aims at shedding light on the world of probiotics, a continuous story of astonishing success in various fields, in particular, the biomedical sector and pharmaceutical industry, as well as to display the importance of probiotics and their therapeutic potential in purpose to compete for sturdy pathogens and to struggle against diseases and acute infections. Shadows and future trends of probiotics use are also discussed.
Collapse
Affiliation(s)
- Mohamed Zommiti
- Unité de Protéomique Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université Tunis El-Manar, Tunis 1006, Tunisia
| | - Marc G. J. Feuilloley
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Université de Rouen Normandie, Normandie Université, F-27000 Evreux, France; (M.G.J.F.); (N.C.)
| | - Nathalie Connil
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Université de Rouen Normandie, Normandie Université, F-27000 Evreux, France; (M.G.J.F.); (N.C.)
| |
Collapse
|
21
|
Liu L, Gu H, Van Limbergen J, Kenney T. SuRF: A new method for sparse variable selection, with application in microbiome data analysis. Stat Med 2020; 40:897-919. [PMID: 33219557 DOI: 10.1002/sim.8809] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 01/16/2023]
Abstract
In this article, we present a new variable selection method for regression and classification purposes, particularly for microbiome analysis. Our method, called subsampling ranking forward selection (SuRF), is based on LASSO penalized regression, subsampling and forward-selection methods. SuRF offers major advantages over existing variable selection methods in terms of both sparsity of selected models and model inference. We provide an R package that can implement our method for generalized linear models. We apply our method to classification problems from microbiome data, using a novel agglomeration approach to deal with the special tree-like correlation structure of the variables. Existing methods arbitrarily choose a taxonomic level a priori before performing the analysis, whereas by combining SuRF with these aggregated variables, we are able to identify the key biomarkers at the appropriate taxonomic level, as suggested by the data. We present simulations in multiple sparse settings to demonstrate that our approach performs better than several other popularly used existing approaches in recovering the true variables. We apply SuRF to two microbiome datasets: one about prediction of pouchitis and another for identifying samples from two healthy individuals. We find that SuRF can provide a better or comparable prediction with other methods while controlling the false positive rate of variable selection.
Collapse
Affiliation(s)
- Lihui Liu
- Department of Mathematics and Statistics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hong Gu
- Department of Mathematics and Statistics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Johan Van Limbergen
- Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Toby Kenney
- Department of Mathematics and Statistics, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
22
|
Metagenome-wide association analysis identifies microbial determinants of post-antibiotic ecological recovery in the gut. Nat Ecol Evol 2020; 4:1256-1267. [PMID: 32632261 DOI: 10.1038/s41559-020-1236-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/28/2020] [Indexed: 12/13/2022]
Abstract
Loss of diversity in the gut microbiome can persist for extended periods after antibiotic treatment, impacting microbiome function, antimicrobial resistance and probably host health. Despite widespread antibiotic use, our understanding of the species and metabolic functions contributing to gut microbiome recovery is limited. Using data from 4 discovery cohorts in 3 continents comprising >500 microbiome profiles from 117 individuals, we identified 21 bacterial species exhibiting robust association with ecological recovery post antibiotic therapy. Functional and growth-rate analysis showed that recovery is supported by enrichment in specific carbohydrate-degradation and energy-production pathways. Association rule mining on 782 microbiome profiles from the MEDUSA database enabled reconstruction of the gut microbial 'food web', identifying many recovery-associated bacteria as keystone species, with the ability to use host- and diet-derived energy sources, and support repopulation of other gut species. Experiments in a mouse model recapitulated the ability of recovery-associated bacteria (Bacteroides thetaiotaomicron and Bifidobacterium adolescentis) to promote recovery with synergistic effects, providing a boost of two orders of magnitude to microbial abundance in early time points and faster maturation of microbial diversity. The identification of specific species and metabolic functions promoting recovery opens up opportunities for rationally determining pre- and probiotic formulations offering protection from long-term consequences of frequent antibiotic usage.
Collapse
|
23
|
Fu J, Zhang Y, Hu Y, Zhao G, Tang Y, Zou L. Concise review: Coarse cereals exert multiple beneficial effects on human health. Food Chem 2020; 325:126761. [PMID: 32387947 DOI: 10.1016/j.foodchem.2020.126761] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/23/2020] [Accepted: 04/05/2020] [Indexed: 02/08/2023]
Abstract
Coarse cereals (CC) refer to cereal grains except for rice and wheat which are highly-valued as functional foods with nutritional and pharmacological properties. Owing to their diverse positive effect on chronic diseases, coarse cereals exert a vital role in food industry. CC and the main contents prevent tumor pathogenesis through promoting apoptosis, inducing cell cycle arrest as well as modulating metastasis initiation. Meanwhile, CC ameliorates cardiovascular diseases through affecting multiple pathways, such as CaMKII/p-BFAF-3, NF-κB, MAPK, PI3K/Akt, etc. Besides, CC and the main contents have potential as prebiotics which facilitating the activities and growth of probiotics such as Bifidobacteria and Lactobacillus. However, there's a lack of report on CC' beneficial properties and the underlying mechanisms are not fully understood. Here this article explains in detail, the effect and mechanism of CC on chronic diseases like tumor, inflammation and cardiovascular diseases.
Collapse
Affiliation(s)
- Jia Fu
- School of Medicine, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China
| | - Yan Zhang
- School of Medicine, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China
| | - Yichen Hu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China
| | - Gang Zhao
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China
| | - Yong Tang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, No.1166, Liutai Avenue, Wenjiang District, Chengdu 611137, Sichuan, China.
| | - Liang Zou
- School of Medicine, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China.
| |
Collapse
|
24
|
Zhou CH, Meng YT, Xu JJ, Fang X, Zhao JL, Zhou W, Zhao J, Han JC, Zhang L, Wang KX, Hu LH, Liao Z, Zou WB, Li ZS, Zou DW. Altered diversity and composition of gut microbiota in Chinese patients with chronic pancreatitis. Pancreatology 2020; 20:16-24. [PMID: 31806503 DOI: 10.1016/j.pan.2019.11.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 11/14/2019] [Accepted: 11/23/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND/OBJECTIVES Gut microbiota alterations in chronic pancreatitis (CP) are seldomly described systematically. It is unknown whether pancreatic exocrine insufficiency (PEI) and different etiologies in patients with CP are associated with gut microbiota dysbiosis. METHODS The fecal microbiota of 69 healthy controls (HCs) and 71 patients with CP were compared to investigate gut microbiome alterations in CP and the relationship among gut microbiome dysbiosis, PEI and different etiologies. Fecal microbiomes were analyzed through 16S ribosomal RNA gene profiling, based on next-generation sequencing. Pancreatic exocrine function was evaluated by determining fecal elastase 1 activity. RESULTS Patients with CP showed gut microbiota dysbiosis with decreased diversity and richness, and taxa-composition changes. On the phylum level, the gut microbiome of the CP group showed lower Firmicutes and Actinobacteria abundances than the HC group and higher Proteobacteria abundances. The abundances of Escherichia-Shigella and other genera were high in gut microbiomes in the CP group, whereas that of Faecalibacterium was low. Kyoto Encyclopedia of Genes and Genomes pathways (lipopolysaccharide biosynthesis and bacterial invasion of epithelial cells) were predicted to be enriched in the CP group. Among the top 5 phyla and 8 genera (in terms of abundance), only Fusobacteria and Eubacterium rectale group showed significant differences between CP patients, with or without PEI. Correlation analysis showed that Bifidobacterium and Lachnoclostridium correlated positively with fecal elastase 1 (r = 0.2616 and 0.2486, respectively, P < 0.05). CONCLUSIONS The current findings indicate that patients with CP have gut microbiota dysbiosis that is partly affected by pancreatic exocrine function.
Collapse
Affiliation(s)
- Chun-Hua Zhou
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China; Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China
| | - Yu-Ting Meng
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Jia-Jia Xu
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Xue Fang
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Jiu-Long Zhao
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Wei Zhou
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Jianhua Zhao
- Shanghai Majorbio Bio-pharm Technology Co., Ltd., China
| | - Ji-Chen Han
- Shanghai Majorbio Bio-pharm Technology Co., Ltd., China
| | - Ling Zhang
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China
| | - Kai-Xuan Wang
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Liang-Hao Hu
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Zhuan Liao
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Wen-Bin Zou
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China.
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China.
| | - Duo-Wu Zou
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China; Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China.
| |
Collapse
|
25
|
Abstract
The gut microbiome is the natural intestinal inhabitant that has been recognized recently as a major player in the maintenance of human health and the pathophysiology of many diseases. Those commensals produce metabolites that have various effects on host biological functions. Therefore, alterations in the normal composition or diversity of microbiome have been implicated in various diseases, including liver cirrhosis and nonalcoholic fatty liver disease. Moreover, accumulating evidence suggests that progression of dysbiosis can be associated with worsening of liver disease. Here, we review the possible roles for gut microbiota in the development, progression, and complication of liver disease.
Collapse
Affiliation(s)
- Somaya A M Albhaisi
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Jasmohan S Bajaj
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University and McGuire Veterans Affairs Medical Center, Richmond, Virginia
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
26
|
Wang Y, Yang G, You L, Yang J, Feng M, Qiu J, Zhao F, Liu Y, Cao Z, Zheng L, Zhang T, Zhao Y. Role of the microbiome in occurrence, development and treatment of pancreatic cancer. Mol Cancer 2019; 18:173. [PMID: 31785619 PMCID: PMC6885316 DOI: 10.1186/s12943-019-1103-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most lethal malignancies. Recent studies indicated that development of pancreatic cancer may be intimately connected with the microbiome. In this review, we discuss the mechanisms through which microbiomes affect the development of pancreatic cancer, including inflammation and immunomodulation. Potential therapeutic and diagnostic applications of microbiomes are also discussed. For example, microbiomes may serve as diagnostic markers for pancreatic cancer, and may also play an important role in determining the efficacies of treatments such as chemo- and immunotherapies. Future studies will provide additional insights into the various roles of microbiomes in pancreatic cancer.
Collapse
Affiliation(s)
- Yicheng Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Jinshou Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Mengyu Feng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Fangyu Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730 China
| |
Collapse
|
27
|
Effect of probiotics supplementation on glucose and oxidative stress in type 2 diabetes mellitus: a meta-analysis of randomized trials. ACTA ACUST UNITED AC 2019; 27:827-837. [PMID: 31691101 DOI: 10.1007/s40199-019-00302-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 09/18/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE The role of oxidative stress in pathogenesis of diabetes is well established. In addition, an association between gut microbiota and type 2 diabetes mellitus (T2DM) is widely observed in previously published reports. This meta-analysis critically examines the association between gut microbiota, and oxidative stress in T2DM. METHODS A systematic search for clinical trials was performed in PubMed, Web of Science and Scopus web databases up to 1 Jan 2019. Primary search terms include "microbiota", "diabetes", and "oxidative stress". Study was conducted according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guideline. All clinical trials that compared the effects of probiotic supplementations with a control group using end points serum levels of fasting blood sugar (FBS), hemoglobin A1C (HbA1C) and oxidative stress biomarkers were included. Two independent researchers screened the data extracted from the relevant studies. The pooled standardized mean difference (SMD) was estimated using the random or fixed effects model. Heterogeneity among the studies was assessed using Q-test. RESULTS Overall, 13 randomized clinical trials (RCTs) involving 840 subjects with T2DM were included in the meta-analysis. The analysis showed that probiotics intake resulted in significant improvement in serum levels of FBS [SMD: -0.35, 95% CI: (-0.59, -0.12)], total antioxidant status (TAS) [SMD: 0.33, 95% CI: (0.11, 0.55)], total glutathione (GSH) [SMD: 0.41, 95% CI: (0.26, 0.56)] and malondialdehyde (MDA) [SMD: -0.54, 95% CI: (-0.83, -0.26)]. No significant improvement was found in HbA1C [SMD: -0.06, 95% CI:(-0.82, 0.69)], and nitric oxide (NO) [SMD:-0.24, 95% CI:(-1.10, 0.62)] levels. CONCLUSION It seems that gut microbiota can exert beneficial effects in diabetic patients via altering oxidative stress' biomarkers. The beneficial effect of gut microbiota however was modest on FBS and non-significant on HbA1C. These results need to be confirmed by conducting more reliable RCTs. PROSPERO REGISTRATION NUMBER CRD42019134905. Graphical abstract Flow diagram of the study selection process.
Collapse
|
28
|
Zhang T, Li Q, Cheng L, Buch H, Zhang F. Akkermansia muciniphila is a promising probiotic. Microb Biotechnol 2019; 12:1109-1125. [PMID: 31006995 PMCID: PMC6801136 DOI: 10.1111/1751-7915.13410] [Citation(s) in RCA: 456] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 12/12/2022] Open
Abstract
Akkermansia muciniphila (A. muciniphila), an intestinal symbiont colonizing in the mucosal layer, is considered to be a promising candidate as probiotics. A. muciniphila is known to have an important value in improving the host metabolic functions and immune responses. Moreover, A. muciniphila may have a value in modifying cancer treatment. However, most of the current researches focus on the correlation between A. muciniphila and diseases, and little is known about the causal relationship between them. Few intervention studies on A. muciniphila are limited to animal experiments, and limited studies have explored its safety and efficacy in humans. Therefore, a critical analysis of the current knowledge in A. muciniphila will play an important foundation for it to be defined as a new beneficial microbe. This article will review the bacteriological characteristics and safety of A. muciniphila, as well as its causal relationship with metabolic disorders, immune diseases and cancer therapy.
Collapse
Affiliation(s)
- Ting Zhang
- Medical Center for Digestive Diseasesthe Second Affiliated Hospital of Nanjing Medical UniversityNanjing210011China
- Key Lab of Holistic Integrative EnterologyNanjing Medical UniversityNanjing210011China
| | - Qianqian Li
- Medical Center for Digestive Diseasesthe Second Affiliated Hospital of Nanjing Medical UniversityNanjing210011China
- Key Lab of Holistic Integrative EnterologyNanjing Medical UniversityNanjing210011China
| | - Lei Cheng
- Biogas Institute of Ministry of Agriculture and Rural AffairsChengdu610041China
- Center for Anaerobic Microbial Resources of Sichuan ProvinceChengdu610041China
| | - Heena Buch
- Medical Center for Digestive Diseasesthe Second Affiliated Hospital of Nanjing Medical UniversityNanjing210011China
| | - Faming Zhang
- Medical Center for Digestive Diseasesthe Second Affiliated Hospital of Nanjing Medical UniversityNanjing210011China
- Key Lab of Holistic Integrative EnterologyNanjing Medical UniversityNanjing210011China
| |
Collapse
|
29
|
Probiotic Lactobacillus and Bifidobacterium strains possess safety characteristics, antiviral activities and host adherence factors revealed by genome mining. EPMA J 2019; 10:337-350. [PMID: 31832110 DOI: 10.1007/s13167-019-00184-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 08/11/2019] [Indexed: 02/07/2023]
Abstract
Background Probiotics belonging to Lactobacillus and Bifidobacterium spp. have been exploited for their health benefits in treatment and prevention of many pathological conditions and promoting human health. Recent advances in understanding probiotics-human interaction through microbiome research in the context of various medical conditions suggest their provisional role in preventive, personalized, and predictive medicine. To streamline their application in disease prevention, development of personalized-based treatments, or their use as biomarkers for predictive diagnosis, in vitro screening for strains with potential probiotic properties should be performed. In this work, we aimed to emphasize the probiotic features of four Lactobacillus and two Bifidobacterium probiotic strains which showed antagonistic properties against microbial pathogens. Methods Firstly, cytotoxicity assessment of cell-free preparations from these strains was performed using a baby hamster kidney (BHK) cells and cell viability was measured by means of sulfo-rhodamine B stain. Secondly, Newcastle disease (ND) and infectious bursal disease (IBD) viruses which pose a great threat in infected poultry were used for assessing antiviral activity of probiotics. Thirdly, the genomes of six probiotic strains were used to identify genes encoding host adherence factors that mediate interaction with human tissues. Results Probiotic preparations exhibited insignificant toxicity as indicated by the high survival rate of BHK cells (surviving fraction varied from 0.82 to 0.99) as compared to the untreated control. Cell-free preparations of probiotics mixed with equal volume of ND and IBD viruses (106 and 104 Tissue Culture Infectious Dose 50, respectively) reduced the titer of ND and IBD viruses on chicken embryo fibroblast cells. Genome mining analysis revealed that the draft genomes of these strains were predicted to encode LPXTG-containing proteins, surface layer proteins, tight adherence pili, sortase-dependent pili, fibronectin, or collagen binding proteins and other factors that adhere to human tissues such as mucus. Such adherence factors enable probiotic bacteria to interact and colonize the host. Conclusion Taken together, safety privileges, antiviral activities, and genomically encoded host interaction factors confirmed probiotic features of the six probiotic strains and their potential in promoting human health.
Collapse
|
30
|
Xiao X, Long W, Huang T, Xia T, Ye R, Liu Y, Long H. Differences Between the Intestinal Lumen Microbiota of Aberrant Crypt Foci (ACF)-Bearing and Non-bearing Rats. Dig Dis Sci 2018; 63:2923-2929. [PMID: 30014223 DOI: 10.1007/s10620-018-5180-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 06/22/2018] [Indexed: 01/19/2023]
Abstract
BACKGROUND Multiple factors including host-microbiota interaction could contribute to the conversion of healthy mucosa to sporadic precancerous lesions. An imbalance of the gut microbiota may be a cause or consequence of this process. AIM The goal was to investigate and analyze the composition of gut microbiota during the genesis of precancerous lesions of colorectal cancer. METHODS To analyze the composition of gut microbiota in the genesis of precancerous lesions, a rat model of 1, 2-dimethylhydrazine (DMH)-induced aberrant crypt foci (ACF) was established. The feces of these rats and healthy rats were collected for 16S rRNA sequencing. RESULTS The diversity and density of the rat intestinal microbiota were significantly different between ACF-bearing and non-bearing group. ACF were induced in rats treated with DMH and showed increased expression of the inflammatory cytokines IL-6, IL-8, and TNF-α. Firmicutes was the most predominant phylum in both ACF-bearing and non-bearing group, followed by Bacteroidetes. Interestingly, although the density of Bacteroidetes decreased from the fifth week to the 17th week in both groups, it was significantly reduced in ACF-bearing group at the 13th week (P < 0.01). At the genus level, no significant difference was observed in the most predominant genus, Lactobacillus. Instead, Bacteroides and Prevotella were significantly less abundant (P < 0.01), while Akkermansia was significantly more abundant (P < 0.05) in ACF-bearing group at the 13th week. CONCLUSION Imbalance of the intestinal microbiota existed between ACF-bearing and non-bearing rats, which could be used as biomarker to predict the genesis of precancerous lesions in the gut.
Collapse
Affiliation(s)
- Xiuli Xiao
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Wenbo Long
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Tingyu Huang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pathology, The First People's Hospital of Neijiang, Neijiang, 641000, Sichuan, China
| | - Tian Xia
- Department of Pathology, The First People's Hospital of Neijiang, Neijiang, 641000, Sichuan, China
| | - Rupei Ye
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yong Liu
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Hanan Long
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Science and Technology, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
31
|
Jayachandran M, Chen J, Chung SSM, Xu B. A critical review on the impacts of β-glucans on gut microbiota and human health. J Nutr Biochem 2018; 61:101-110. [DOI: 10.1016/j.jnutbio.2018.06.010] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 06/04/2018] [Accepted: 06/26/2018] [Indexed: 02/07/2023]
|
32
|
Pancreatic Cancer and Obesity: Molecular Mechanisms of Cell Transformation and Chemoresistance. Int J Mol Sci 2018; 19:ijms19113331. [PMID: 30366466 PMCID: PMC6274743 DOI: 10.3390/ijms19113331] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/27/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022] Open
Abstract
Cancer and obesity are the two major epidemics of the 21st century. Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of death, with a five-year overall survival rate of only 8%. Its incidence and mortality have increased in recent years, and this cancer type is expected to be among the top five leading causes of cancer-related death by 2030 in the United States (US). In the last three decades, the prevalence of overweight people has boosted with a consequent increase in obesity-related diseases. Considerable epidemiologic evidence correlates overweight and obese conditions to an increased risk of several types of cancer, including PDAC. Besides being a risk factor for multiple metabolic disorders, the tumor-promoting effects of obesity occur at the local level via inflammatory mediators that are associated with adipose inflammation and metabolic or hormones mediators and microbiota dysbiosis. Although an excess of body mass index (BMI) represents the second most modifiable risk factor for PDAC with an increased cancer related-death of more than 20–40%, still little is known about the molecular mechanisms that underlie this strong association. In this review, we focused on the role of obesity as a preventable risk factor of PDAC, discussing the molecular mechanisms linking obesity to cancer initiation and progression. Moreover, we highlighted the role of obesity in defining chemoresistance, showing how a high BMI can actually reduce response to chemotherapy.
Collapse
|
33
|
The suppression of TXNIP and miR-200c improve beta-cell function in patients with Type 2 diabetes: A randomized, double-blind, placebo-controlled trial. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.07.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
34
|
Kim S, Huang E, Park S, Holzapfel W, Lim SD. Physiological Characteristics and Anti-obesity Effect of Lactobacillus plantarum K10. Korean J Food Sci Anim Resour 2018; 38:554-569. [PMID: 30018499 PMCID: PMC6048381 DOI: 10.5851/kosfa.2018.38.3.554] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 11/25/2022] Open
Abstract
This study aimed to investigate the physiological characteristics and anti-obesity effects of Lactobacillus plantarum K10. The α-amylase inhibitory activity, α-glucosidase inhibitory activity, and lipase inhibitory activity of L. plantarum K10 was 94.66±4.34%, 99.78±0.12%, and 87.40±1.41%, respectively. Moreover, the strain inhibited the adipocyte differentiation of 3T3-L1 cells (32.61±8.32%) at a concentration of 100 µg/mL. In order to determine its potential for use as a probiotic, we investigated the physiological characteristics of L. plantarum K10. L. plantarum K10 was resistant to gentamycin, kanamycin, streptomycin, ampicillin, ciprofloxacin, tetracycline, vancomycin, and chloramphenicol. It also showed higher Leucine arylamidase, Valine arylamidase, and β-galactosidase activities. Moreover, it was comparatively tolerant to bile juice and acid, exhibiting resistance to Escherichia coli, Salmonella Typhimurium, Listeria monocytogenes, and Staphylococcus aureus with rates of 90.71%, 11.86%, 14.19%, and 23.08%, respectively. The strain did not produce biogenic amines and showed higher adhesion to HT-29 cells compared to L. rhamnosus GG. As a result of the animal study, L. plantarum K10 showed significantly lower body weight compared to the high-fat diet group. The administration of L. plantarum K10 resulted in a reduction of subcutaneous fat mass and mesenteric fat mass compared to the high-fat diet (HFD) group. L. plantarum K10 also showed improvement in gut permeability compared to the HFD positive control group. These results demonstrate that L. plantarum K10 has potential as a probiotic with anti-obesity effects.
Collapse
Affiliation(s)
- Seulki Kim
- Korea Food Research Institute,
Wanju 55365, Korea
| | - Eunchong Huang
- Advanced Green Energy and Environment, Handong
Global University, Pohang 37673,
Korea
| | - Soyoung Park
- Advanced Green Energy and Environment, Handong
Global University, Pohang 37673,
Korea
| | - Wilhelm Holzapfel
- Advanced Green Energy and Environment, Handong
Global University, Pohang 37673,
Korea
- Holzapfel Effective Microbes,
103 Techno-Business Center, Pohang 37673, Korea
| | | |
Collapse
|
35
|
Deaver JA, Eum SY, Toborek M. Circadian Disruption Changes Gut Microbiome Taxa and Functional Gene Composition. Front Microbiol 2018; 9:737. [PMID: 29706947 PMCID: PMC5909328 DOI: 10.3389/fmicb.2018.00737] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 03/29/2018] [Indexed: 12/22/2022] Open
Abstract
Disrupted circadian rhythms and alterations of the gut microbiome composition were proposed to affect host health. Therefore, the aim of this research was to identify whether these events are connected and if circadian rhythm disruption by abnormal light–dark (LD) cycles affects microbial community gene expression and host vulnerability to intestinal dysfunction. Mice were subjected to either a 4-week period of constant 24-h light or of normal 12-h LD cycles. Stool samples were collected at the beginning and after the circadian rhythm disruption. A metatranscriptomic analysis revealed an increase in Ruminococcus torques, a bacterial species known to decrease gut barrier integrity, and a decrease in Lactobacillus johnsonii, a bacterium that helps maintain the intestinal epithelial cell layer, after circadian rhythm disruption. In addition, genes involved in pathways promoting host beneficial immune responses were downregulated, while genes involved in the synthesis and transportation of the endotoxin lipopolysaccharide were upregulated in mice with disrupted circadian cycles. Importantly, these mice were also more prone to dysfunction of the intestinal barrier. These results further elucidate the impact of light-cycle disruption on the gut microbiome and its connection with increased incidence of disease in response to circadian rhythm disturbances.
Collapse
Affiliation(s)
- Jessica A Deaver
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sung Y Eum
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
36
|
Vlasova AN, Rajashekara G, Saif LJ. Interactions between human microbiome, diet, enteric viruses and immune system: Novel insights from gnotobiotic pig research. ACTA ACUST UNITED AC 2018; 28:95-103. [PMID: 33149747 PMCID: PMC7594741 DOI: 10.1016/j.ddmod.2019.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Studies over the past few decades demonstrated that gnotobiotic (Gn) pigs provide an unprecedented translational model to study human intestinal health and diseases. Due to the high degree of anatomical, physiological, metabolic, immunological, and developmental similarity, the domestic pig closely mimics the human intestinal microenvironment. Also, Gn piglets can be efficiently transplanted with human microbiota from infants, children and adults with resultant microbial profiles remarkably similar to the original human samples, a feat consistently not achievable in rodent models. Finally, Gn and human microbiota-associated (HMA) piglets are susceptible to human enteric viral pathogens (including human rotavirus, HRV) and can be fed authentic human diets, which further increases the translational potential of these models. In this review, we will focus on recent studies that evaluated the pathophysiology of protein malnutrition and the associated dysbiosis and immunological dysfunction in neonatal HMA piglets. Additionally, we will discuss studies of potential dietary interventions that moderate the effects of malnutrition and dysbiosis on antiviral immunity and HRV vaccines in HMA pigs. Such studies provide novel models and novel mechanistic insights critical for development of drug interventions.
Collapse
Affiliation(s)
- Anastasia N Vlasova
- Food Animal Health Research Program, CFAES, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Gireesh Rajashekara
- Food Animal Health Research Program, CFAES, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Linda J Saif
- Food Animal Health Research Program, CFAES, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| |
Collapse
|
37
|
Ghoddusi H, Thomas L. Microbiota of the Human Gut. PROBIOTIC DAIRY PRODUCTS 2017:1-15. [DOI: 10.1002/9781119214137.ch1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
38
|
Zoumpopoulou G, Tsakalidou E, Thomas L. An Overview of Probiotic Research. PROBIOTIC DAIRY PRODUCTS 2017:293-357. [DOI: 10.1002/9781119214137.ch8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
39
|
Perinatal Bisphenol A Exposure Induces Chronic Inflammation in Rabbit Offspring via Modulation of Gut Bacteria and Their Metabolites. mSystems 2017; 2:mSystems00093-17. [PMID: 29034330 PMCID: PMC5634791 DOI: 10.1128/msystems.00093-17] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/17/2017] [Indexed: 02/06/2023] Open
Abstract
Emerging evidence suggests that environmental toxicants may influence inflammation-promoted chronic disease susceptibility during early life. BPA, an environmental endocrine disruptor, can transfer across the placenta and accumulate in fetal gut and liver. However, underlying mechanisms for BPA-induced colonic and liver inflammation are not fully elucidated. In this report, we show how perinatal BPA exposure in rabbits alters gut microbiota and their metabolite profiles, which leads to colonic and liver inflammation as well as to increased gut permeability as measured by elevated serum lipopolysaccharide (LPS) levels in the offspring. Also, perinatal BPA exposure leads to reduced levels of gut bacterial diversity and bacterial metabolites (short-chain fatty acids [SCFA]) and elevated gut permeability—three common early biomarkers of inflammation-promoted chronic diseases. In addition, we showed that SCFA ameliorated BPA-induced intestinal permeability in vitro. Thus, our study results suggest that correcting environmental toxicant-induced bacterial dysbiosis early in life may reduce the risk of chronic diseases later in life. Bisphenol A (BPA) accumulates in the maturing gut and liver in utero and is known to alter gut bacterial profiles in offspring. Gut bacterial dysbiosis may contribute to chronic colonic and systemic inflammation. We hypothesized that perinatal BPA exposure-induced intestinal (and liver) inflammation in offspring is due to alterations in the microbiome and colonic metabolome. The 16S rRNA amplicon sequencing analysis revealed differences in beta diversity with a significant reduction in the relative abundances of short-chain fatty acid (SCFA) producers such as Oscillospira and Ruminococcaceae due to BPA exposure. Furthermore, BPA exposure reduced fecal SCFA levels and increased systemic lipopolysaccharide (LPS) levels. BPA exposure-increased intestinal permeability was ameliorated by the addition of SCFA in vitro. Metabolic fingerprints revealed alterations in global metabolism and amino acid metabolism. Thus, our findings indicate that perinatal BPA exposure may cause gut bacterial dysbiosis and altered metabolite profiles, particularly SCFA profiles, leading to chronic colon and liver inflammation. IMPORTANCE Emerging evidence suggests that environmental toxicants may influence inflammation-promoted chronic disease susceptibility during early life. BPA, an environmental endocrine disruptor, can transfer across the placenta and accumulate in fetal gut and liver. However, underlying mechanisms for BPA-induced colonic and liver inflammation are not fully elucidated. In this report, we show how perinatal BPA exposure in rabbits alters gut microbiota and their metabolite profiles, which leads to colonic and liver inflammation as well as to increased gut permeability as measured by elevated serum lipopolysaccharide (LPS) levels in the offspring. Also, perinatal BPA exposure leads to reduced levels of gut bacterial diversity and bacterial metabolites (short-chain fatty acids [SCFA]) and elevated gut permeability—three common early biomarkers of inflammation-promoted chronic diseases. In addition, we showed that SCFA ameliorated BPA-induced intestinal permeability in vitro. Thus, our study results suggest that correcting environmental toxicant-induced bacterial dysbiosis early in life may reduce the risk of chronic diseases later in life.
Collapse
|
40
|
Probiotics as an adjunct for the treatment of recurrent wheezing in infants and effects on expression of T-helper 1 and regulatory T cytokines. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.06.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
41
|
|
42
|
Zheng M, Zhang R, Tian X, Zhou X, Pan X, Wong A. Assessing the Risk of Probiotic Dietary Supplements in the Context of Antibiotic Resistance. Front Microbiol 2017; 8:908. [PMID: 28579981 PMCID: PMC5437161 DOI: 10.3389/fmicb.2017.00908] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/04/2017] [Indexed: 11/13/2022] Open
Abstract
Probiotic bacteria are known to harbor intrinsic and mobile genetic elements that confer resistance to a wide variety of antibiotics. Their high amounts in dietary supplements can establish a reservoir of antibiotic resistant genes in the human gut. These resistant genes can be transferred to pathogens that share the same intestinal habitat thus resulting in serious clinical ramifications. While antibiotic resistance of probiotic bacteria from food, human and animal sources have been well-documented, the resistant profiles of probiotics from dietary supplements have only been recently studied. These products are consumed with increasing regularity due to their health claims that include the improvement of intestinal health and immune response as well as prevention of acute and antibiotic-associated diarrhea and cancer; but, a comprehensive risk assessment on the spread of resistant genes to human health is lacking. Here, we highlight recent reports of antibiotic resistance of probiotic bacteria isolated from dietary supplements, and propose complementary strategies that can shed light on the risks of consuming such products in the context of a global widespread of antibiotic resistance. In concomitant with a broader screening of antibiotic resistance in probiotic supplements is the use of computational simulations, live imaging and functional genomics to harvest knowledge on the evolutionary behavior, adaptations and dynamics of probiotics studied in conditions that best represent the human gut including in the presence of antibiotics. The underlying goal is to enable the health benefits of probiotics to be exploited in a responsible manner and with minimal risk to human health.
Collapse
Affiliation(s)
- Min Zheng
- College of Natural, Applied and Health Sciences, Wenzhou-Kean UniversityWenzhou, China
| | - Ruijia Zhang
- College of Natural, Applied and Health Sciences, Wenzhou-Kean UniversityWenzhou, China
| | - Xuechen Tian
- College of Natural, Applied and Health Sciences, Wenzhou-Kean UniversityWenzhou, China
| | - Xuan Zhou
- College of Natural, Applied and Health Sciences, Wenzhou-Kean UniversityWenzhou, China
| | - Xutong Pan
- College of Natural, Applied and Health Sciences, Wenzhou-Kean UniversityWenzhou, China
| | - Aloysius Wong
- College of Natural, Applied and Health Sciences, Wenzhou-Kean UniversityWenzhou, China
| |
Collapse
|
43
|
Dror T, Dickstein Y, Dubourg G, Paul M. Microbiota manipulation for weight change. Microb Pathog 2017; 106:146-161. [PMID: 26792677 DOI: 10.1016/j.micpath.2016.01.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 12/21/2022]
Abstract
Manipulation of the intestinal microbiota has been linked to weight changes and obesity. To explore the influence of specific agents that alter the intestinal flora on weight in different patient groups we conducted a meta-analysis of randomized controlled trials (RCTs) reporting on the effects of probiotics, prebiotics, synbiotics, and antibiotics on weight. We searched the Pubmed and Cochrane Library databases for trials on adults, children, and infants evaluating the effects of these substances on weight. Our primary outcome was weight change from baseline. Standardized mean differences (SMDs) with 95% confidence intervals were calculated. We identified and included 13 adult, 17 children, and 23 infant RCTs. Effects were opposite among adults and children, showing weight loss among adults (SMD -0.54 [-0.83, -0.25)) and minor weight gains among children (SMD 0.20 [0.04, 0.36]) and infants (SMD 0.30 [-0.01, 0.62]) taking mainly Lactobacillus probiotic supplements. Heterogeneity was substantial in the adult and infant analyses and could not be explained by intervention or patient characteristics. Azithromycin administration in children with pulmonary disease was associated with weight gain (SMD 0.39 [0.24, 0.54]), without heterogeneity. A high risk of selective reporting and attrition bias was detected across the studies, making it difficult to draw firm conclusions. Overall, our meta-analysis suggests that there may be a role for probiotics in promoting weight loss in adults and weight gain in children, however additional studies are needed. Though we cannot recommend antibiotic administration for weight manipulation, its use provides advantageous weight gain in children with cystic fibrosis and bronchiectasis.
Collapse
Affiliation(s)
- Tal Dror
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Yaakov Dickstein
- Infectious Diseases Institute, Rambam Health Care Campus, Haifa, Israel
| | - Grégory Dubourg
- Pôle des Maladies Infectieuses et Tropicales Clinique et Biologique, Fédération de Bactériologie-Hygiène-Virologie, University, Hospital Centre Timone, Institut Hospitalo-Universitaire (IHU) Méditerranée Infection, Assistance Publique - Hôpitaux de Marseille, Marseille, France; Université Aix-Marseille, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes (URMITE) UM 63 CNRS 7278 IRD 198 INSERM U1095, Facultés de Médecine et de Pharmacie, Marseille, France
| | - Mical Paul
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel; Infectious Diseases Institute, Rambam Health Care Campus, Haifa, Israel.
| |
Collapse
|
44
|
Early pregnancy probiotic supplementation with Lactobacillus rhamnosus HN001 may reduce the prevalence of gestational diabetes mellitus: a randomised controlled trial. Br J Nutr 2017; 117:804-813. [PMID: 28367765 PMCID: PMC5426322 DOI: 10.1017/s0007114517000289] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The study aims to assess whether supplementation with the probiotic Lactobacillus rhamnosus HN001 (HN001) can reduce the prevalence of gestational diabetes mellitus (GDM). A double-blind, randomised, placebo-controlled parallel trial was conducted in New Zealand (NZ) (Wellington and Auckland). Pregnant women with a personal or partner history of atopic disease were randomised at 14–16 weeks’ gestation to receive HN001 (6×109 colony-forming units) (n 212) or placebo (n 211) daily. GDM at 24–30 weeks was assessed using the definition of the International Association of Diabetes and Pregnancy Study Groups (IADPSG) (fasting plasma glucose ≥5·1 mmol/l, or 1 h post 75 g glucose level at ≥10 mmol/l or at 2 h ≥8·5 mmol/l) and NZ definition (fasting plasma glucose ≥5·5 mmol/l or 2 h post 75 g glucose at ≥9 mmol/l). All analyses were intention-to-treat. A total of 184 (87 %) women took HN001 and 189 (90 %) women took placebo. There was a trend towards lower relative rates (RR) of GDM (IADPSG definition) in the HN001 group, 0·59 (95 % CI 0·32, 1·08) (P=0·08). HN001 was associated with lower rates of GDM in women aged ≥35 years (RR 0·31; 95 % CI 0·12, 0·81, P=0·009) and women with a history of GDM (RR 0·00; 95 % CI 0·00, 0·66, P=0·004). These rates did not differ significantly from those of women without these characteristics. Using the NZ definition, GDM prevalence was significantly lower in the HN001 group, 2·1 % (95 % CI 0·6, 5·2), v. 6·5 % (95 % CI 3·5, 10·9) in the placebo group (P=0·03). HN001 supplementation from 14 to 16 weeks’ gestation may reduce GDM prevalence, particularly among older women and those with previous GDM.
Collapse
|
45
|
Hoffman DJ, Campos-Ponce M, Taddei CR, Doak CM. Microbiome, growth retardation and metabolism: are they related? Ann Hum Biol 2017; 44:201-207. [PMID: 27927018 DOI: 10.1080/03014460.2016.1267261] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CONTEXT Despite an improvement in food security and the delivery of nutritional supplements to children living in impoverished parts of the world, poor growth is still highly prevalent. Given that the microbiome is related to both nutrient absorption, as well as overweight/obesity, it may play a central role in limiting or modifying normal growth processes while contributing to chronic disease risks. OBJECTIVE The objective of this paper is to describe normal growth processes, the role of the microbiome in supporting or disrupting normal growth processes, and its potential impact on long-term health. METHODS A literature search of relevant human and laboratory research on growth, microbiome and the relationship between poor growth and chronic diseases was conducted. This review focuses on potential mechanisms that implicate the microbiome as a mediator of poor growth and later metabolic outcomes. In this relationship, attention was given to the potential for gastrointestinal infections to disrupt the microbiome. RESULTS Based on the studies reviewed, it is clear that exposure to infections disturbs both intestinal functioning as well as normal growth and changes in the microbiome may influence micronutrient availability and metabolic processes. CONCLUSIONS The microbiome may play a significant role in limiting human growth, but little is known about changes in the microbiome during periods of undernutrition. Thus, it is of great scientific and public health importance to improve the understanding of how the microbiome changes during nutrient deprivation. To best address the issue of the double burden and poor growth in low-income countries, research is warranted to advance the knowledge of the long-term role of the microbiome in the health of children exposed to undernutrition.
Collapse
Affiliation(s)
- Daniel J Hoffman
- a Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition, and Health , Rutgers, the State University of New Jersey , New Brunswick , NJ , USA
| | - Maiza Campos-Ponce
- b Department of Health Sciences , VU University , Amsterdam , The Netherlands
| | - Carla R Taddei
- c Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences , University of São Paulo , São Paulo, SP , Brazil.,d School of Arts, Science and Humanities , University of São Paulo , São Paulo, SP , Brazil
| | - Colleen M Doak
- b Department of Health Sciences , VU University , Amsterdam , The Netherlands
| |
Collapse
|
46
|
Kubelkova K, Benuchova M, Kozakova H, Sinkora M, Krocova Z, Pejchal J, Macela A. Gnotobiotic mouse model's contribution to understanding host-pathogen interactions. Cell Mol Life Sci 2016; 73:3961-9. [PMID: 27544211 PMCID: PMC11108488 DOI: 10.1007/s00018-016-2341-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 07/25/2016] [Accepted: 08/11/2016] [Indexed: 02/07/2023]
Abstract
This brief review is dedicated to the legacy of Prof. Jaroslav Šterzl and his colleagues, who laid the foundation for gnotobiology in the former Czechoslovakia 55 years. Prof. Sterzl became one of the founders of modern Czechoslovak immunology, which was characterized by work on a wide range of problems needing to be solved. While examining the mechanisms of innate immunity, he focused his studies on the induction of antibody production by immunocompetent cells involved in adaptive immune transmission while using the model of pig fetuses and germ-free piglets and characterizing immunoglobulins in the sera of these piglets. Although not fully appreciated to this day, his experimental proof of the hypothesis focused on the common precursor of cell-forming antibodies of different isotypes was later confirmed in experiments at the gene level. Prof. Sterzl's work represented a true milestone in the development of not solely Czechoslovak but also European and global immunology. He collaborated closely with the World Health Organization for many years, serving there as leader of the Reference Laboratory for Factors of Innate Immunity.
Collapse
Affiliation(s)
- Klara Kubelkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 1575 Trebesska, 500 01, Hradec Kralove, Czech Republic.
| | - Milota Benuchova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 1575 Trebesska, 500 01, Hradec Kralove, Czech Republic
| | - Hana Kozakova
- Laboratory of Gnotobiology, Czech Academy of Sciences, Institute of Microbiology, 549 22, Novy Hradek, Czech Republic
| | - Marek Sinkora
- Laboratory of Gnotobiology, Czech Academy of Sciences, Institute of Microbiology, 549 22, Novy Hradek, Czech Republic
| | - Zuzana Krocova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 1575 Trebesska, 500 01, Hradec Kralove, Czech Republic
| | - Jaroslav Pejchal
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, 1575 Trebesska, 500 01, Hradec Kralove, Czech Republic
| | - Ales Macela
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 1575 Trebesska, 500 01, Hradec Kralove, Czech Republic
| |
Collapse
|
47
|
Javurek AB, Spollen WG, Johnson SA, Bivens NJ, Bromert KH, Givan SA, Rosenfeld CS. Effects of exposure to bisphenol A and ethinyl estradiol on the gut microbiota of parents and their offspring in a rodent model. Gut Microbes 2016; 7:471-485. [PMID: 27624382 PMCID: PMC5103659 DOI: 10.1080/19490976.2016.1234657] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gut dysbiosis may result in various diseases, such as metabolic and neurobehavioral disorders. Exposure to endocrine disrupting chemicals (EDCs), including bisphenol A (BPA) and ethinyl estradiol (EE), especially during development, may also increase the risk for such disorders. An unexplored possibility is that EDC-exposure might alter the gut microbial composition. Gut flora and their products may thus be mediating factors for the disease-causing effects of these chemicals. To examine the effects of EDCs on the gut microbiome, female and male monogamous and biparental California mice (Peromyscus californicus) were exposed to BPA (50 mg/kg feed weight) or EE (0.1 ppb) or control diet from periconception through weaning. 16s rRNA sequencing was performed on bacterial DNA isolated from fecal samples, and analyses performed for P0 and F1 males and females. Both BPA and EE induced generational and sex-dependent gut microbiome changes. Many of the bacteria, e.g. Bacteroides, Mollicutes, Prevotellaceae, Erysipelotrichaceae, Akkermansia, Methanobrevibacter, Sutterella, whose proportions increase with exposure to BPA or EE in the P0 or F1 generation are associated with different disorders, such as inflammatory bowel disease (IBD), metabolic disorders, and colorectal cancer. However, the proportion of the beneficial bacterium, Bifidobacterium, was also elevated in fecal samples of BPA- and EE-exposed F1 females. Intestinal flora alterations were also linked to changes in various metabolic and other pathways. Thus, BPA and EE exposure may disrupt the normal gut flora, which may in turn result in systemic effects. Probiotic supplementation might be an effective means to mitigate disease-promoting effects of these chemicals.
Collapse
Affiliation(s)
- Angela B. Javurek
- Bond Life Sciences Center, University of Missouri, Columbia, MO USA,Biomedical Sciences, University of Missouri, Columbia, MO USA
| | - William G. Spollen
- Bond Life Sciences Center, University of Missouri, Columbia, MO USA,Informatics Research Core Facility, University of Missouri, Columbia, MO USA
| | - Sarah A. Johnson
- Bond Life Sciences Center, University of Missouri, Columbia, MO USA,Biomedical Sciences, University of Missouri, Columbia, MO USA,Animal Sciences, University of Missouri, Columbia, MO USA
| | | | | | - Scott A. Givan
- Bond Life Sciences Center, University of Missouri, Columbia, MO USA,Informatics Research Core Facility, University of Missouri, Columbia, MO USA,Molecular Microbiology and Immunology, University of Missouri, Columbia, MO USA
| | - Cheryl S. Rosenfeld
- Bond Life Sciences Center, University of Missouri, Columbia, MO USA,Biomedical Sciences, University of Missouri, Columbia, MO USA,Genetics Area Program, University of Missouri, Columbia, MO USA,Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO USA,CONTACT Cheryl S. Rosenfeld, DVM, PhD Biomedical Sciences and Bond Life Sciences Center, University of Missouri, 440F Bond Life Sciences Center, 1201 E. Rollins Rd., Columbia, MO 65211
| |
Collapse
|
48
|
Oriá RB, Murray-Kolb LE, Scharf RJ, Pendergast LL, Lang DR, Kolling GL, Guerrant RL. Early-life enteric infections: relation between chronic systemic inflammation and poor cognition in children. Nutr Rev 2016; 74:374-86. [PMID: 27142301 DOI: 10.1093/nutrit/nuw008] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The intestinal microbiota undergoes active remodeling in the first 6 to 18 months of life, during which time the characteristics of the adult microbiota are developed. This process is strongly influenced by the early diet and enteric pathogens. Enteric infections and malnutrition early in life may favor microbiota dysbiosis and small intestinal bacterial overgrowth, resulting in intestinal barrier dysfunction and translocation of intestinal bacterial products, ultimately leading to low-grade, chronic, subclinical systemic inflammation. The leaky gut-derived low-grade systemic inflammation may have profound consequences on the gut-liver-brain axis, compromising normal growth, metabolism, and cognitive development. This review examines recent data suggesting that early-life enteric infections that lead to intestinal barrier disruption may shift the intestinal microbiota toward chronic systemic inflammation and subsequent impaired cognitive development.
Collapse
Affiliation(s)
- Reinaldo B Oriá
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA.
| | - Laura E Murray-Kolb
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Rebecca J Scharf
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Laura L Pendergast
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Dennis R Lang
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Glynis L Kolling
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Richard L Guerrant
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
49
|
González-Arancibia C, Escobar-Luna J, Barrera-Bugueño C, Díaz-Zepeda C, González-Toro MP, Olavarría-Ramírez L, Zanelli-Massai F, Gotteland M, Bravo JA, Julio-Pieper M. What goes around comes around: novel pharmacological targets in the gut-brain axis. Therap Adv Gastroenterol 2016; 9:339-53. [PMID: 27134664 PMCID: PMC4830101 DOI: 10.1177/1756283x16630718] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gut and the brain communicate bidirectionally through anatomic and humoral pathways, establishing what is known as the gut-brain axis. Therefore, interventions affecting one system will impact on the other, giving the opportunity to investigate and develop future therapeutic strategies that target both systems. Alterations in the gut-brain axis may arise as a consequence of changes in microbiota composition (dysbiosis), modifications in intestinal barrier function, impairment of enteric nervous system, unbalanced local immune response and exaggerated responses to stress, to mention a few. In this review we analyze and discuss several novel pharmacological targets within the gut-brain axis, with potential applications to improve intestinal and mental health.
Collapse
Affiliation(s)
- Camila González-Arancibia
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Jorge Escobar-Luna
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Camila Barrera-Bugueño
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Camilo Díaz-Zepeda
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - María P. González-Toro
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Loreto Olavarría-Ramírez
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Francesca Zanelli-Massai
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Martin Gotteland
- Departamento de Nutrición, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Javier A. Bravo
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | | |
Collapse
|
50
|
Abstract
Gnotobiotic (GN) rodent models have provided insight into the contributions of the gut microbiota to host health and preventing disease. However, rodent models are limited by several important physiological and metabolic differences from humans, and many rodent models do not dependably replicate the clinical manifestations of human diseases. Due to the high degree of similarity in anatomy, physiology, immunology and brain growth, the domestic pig (Sus scrofa) is considered a clinically relevant model to study factors influencing human gastrointestinal, immune, and brain development. Gnotobiotic piglet models have been developed and shown to recapitulate key aspects of GN rodent models. Human microbiota-associated (HMA) piglets have been established using inocula from infants, children, and adults. The gut microbiota of recipient HMA piglets was more similar to that of the human donor than that of conventionally reared piglets harboring a pig microbiota. Moreover, Bifidobacterium and Bacteroides, two predominant bacterial groups of infant gut, were successfully established in the HMA piglets. Thus, the HMA pig model has the potential to be a valuable model for investigating how the gut microbiota composition changes in response to environmental factors, such as age, diet, vaccination, antibiotic use and infection. The HMA also represents a robust model for screening the efficacy of pre- and probiotic interventions. Lastly, HMA piglets can be an ideal model with which to elucidate microbe-host interactions in human health and disease due to the similarities to humans in anatomy, physiology, developmental maturity at birth, and the pathophysiology of many human diseases.
Collapse
Affiliation(s)
- Mei Wang
- Mei Wang, PhD, is a research specialist and Sharon M. Donovan, PhD, RD, is a professor in the Department of Food Science & Human Nutrition, University of Illinois, Urbana, Illinois
| | - Sharon M Donovan
- Mei Wang, PhD, is a research specialist and Sharon M. Donovan, PhD, RD, is a professor in the Department of Food Science & Human Nutrition, University of Illinois, Urbana, Illinois
| |
Collapse
|