101
|
Kennedy KM, de Goffau MC, Perez-Muñoz ME, Arrieta MC, Bäckhed F, Bork P, Braun T, Bushman FD, Dore J, de Vos WM, Earl AM, Eisen JA, Elovitz MA, Ganal-Vonarburg SC, Gänzle MG, Garrett WS, Hall LJ, Hornef MW, Huttenhower C, Konnikova L, Lebeer S, Macpherson AJ, Massey RC, McHardy AC, Koren O, Lawley TD, Ley RE, O'Mahony L, O'Toole PW, Pamer EG, Parkhill J, Raes J, Rattei T, Salonen A, Segal E, Segata N, Shanahan F, Sloboda DM, Smith GCS, Sokol H, Spector TD, Surette MG, Tannock GW, Walker AW, Yassour M, Walter J. Questioning the fetal microbiome illustrates pitfalls of low-biomass microbial studies. Nature 2023; 613:639-649. [PMID: 36697862 PMCID: PMC11333990 DOI: 10.1038/s41586-022-05546-8] [Citation(s) in RCA: 181] [Impact Index Per Article: 90.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/09/2022] [Indexed: 01/26/2023]
Abstract
Whether the human fetus and the prenatal intrauterine environment (amniotic fluid and placenta) are stably colonized by microbial communities in a healthy pregnancy remains a subject of debate. Here we evaluate recent studies that characterized microbial populations in human fetuses from the perspectives of reproductive biology, microbial ecology, bioinformatics, immunology, clinical microbiology and gnotobiology, and assess possible mechanisms by which the fetus might interact with microorganisms. Our analysis indicates that the detected microbial signals are likely the result of contamination during the clinical procedures to obtain fetal samples or during DNA extraction and DNA sequencing. Furthermore, the existence of live and replicating microbial populations in healthy fetal tissues is not compatible with fundamental concepts of immunology, clinical microbiology and the derivation of germ-free mammals. These conclusions are important to our understanding of human immune development and illustrate common pitfalls in the microbial analyses of many other low-biomass environments. The pursuit of a fetal microbiome serves as a cautionary example of the challenges of sequence-based microbiome studies when biomass is low or absent, and emphasizes the need for a trans-disciplinary approach that goes beyond contamination controls by also incorporating biological, ecological and mechanistic concepts.
Collapse
Affiliation(s)
- Katherine M Kennedy
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Marcus C de Goffau
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Vascular Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Wellcome Sanger Institute, Cambridge, UK
| | - Maria Elisa Perez-Muñoz
- Department of Agriculture, Food and Nutrition Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Marie-Claire Arrieta
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany
- Yonsei Frontier Lab (YFL), Yonsei University, Seoul, South Korea
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Thorsten Braun
- Department of Obstetrics and Experimental Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frederic D Bushman
- Department of Microbiology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel Dore
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS, Jouy-en-Josas, France
| | - Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Ashlee M Earl
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Jonathan A Eisen
- Department of Evolution and Ecology, University of California, Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- UC Davis Genome Center, University of California, Davis, Davis, CA, USA
| | - Michal A Elovitz
- Maternal and Child Health Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Stephanie C Ganal-Vonarburg
- Universitätsklinik für Viszerale Chirurgie und Medizin, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Michael G Gänzle
- Department of Agriculture, Food and Nutrition Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
- Department of Medicine and Division of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Lindsay J Hall
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
- Chair of Intestinal Microbiome, ZIEL-Institute for Food and Health, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Curtis Huttenhower
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Liza Konnikova
- Departments of Pediatrics and Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Andrew J Macpherson
- Department for Biomedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Ruth C Massey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Alice Carolyn McHardy
- Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), Hannover Braunschweig site, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Trevor D Lawley
- Department of Vascular Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Liam O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Paul W O'Toole
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Eric G Pamer
- Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Julian Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Jeroen Raes
- VIB Center for Microbiology, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Thomas Rattei
- Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eran Segal
- Weizmann Institute of Science, Rehovot, Israel
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Fergus Shanahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Harry Sokol
- Gastroenterology Department, AP-HP, Saint Antoine Hospital, Centre de Recherche Saint-Antoine, CRSA, INSERM and Sorbonne Université, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - Tim D Spector
- Department of Twin Research, King's College London, London, UK
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gerald W Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Alan W Walker
- Gut Health Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Moran Yassour
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jens Walter
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- School of Microbiology, University College Cork, Cork, Ireland.
- Department of Medicine, University College Cork, Cork, Ireland.
| |
Collapse
|
102
|
Analysis of Scat for Gut Microbiome Identification in Wolves from a Mediterranean and an Alpine Area. DIVERSITY 2022. [DOI: 10.3390/d15010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The gut microbiome can play a fundamental role in several processes associated with an organism’s ecology, and research on the microbiota of wild animals has flourished in the last decades. Microbiome composition can vary across and within species according to taxonomy and environmental variability, including the availability of food resources. Species with a large distribution may exhibit spatial patterns acting at local/regional scales. We considered one of the most widespread and ecologically important predators in the world, i.e., the grey wolf Canis lupus, for which microbiome data is unduly limited. We studied four packs in different ecological conditions in Italy—two packs from a Mediterranean coastal area and two packs from an Alpine range—using an amplicon sequencing barcoding approach. Overall, our results are consistent with food habits entailing a diet largely based on wild prey and agree with findings obtained on other species of canids. If confirmed through a larger sample, they would support the hypothesis of an influence of the shared evolutionary history across canids on the composition of the gut microbiome. Some emerging differences were observed among packs in terms of species composition (Jaccard) and diversity, providing partial support to recent indications on pack identity as a significant determinant of microbiome composition. These results should be considered preliminary results of gut microbiome composition in our study areas.
Collapse
|
103
|
Dualib PM, Fernandes G, Taddei CR, Carvalho CRS, Sparvoli LG, Bittencourt C, Silva IT, Mattar R, Ferreira SRG, Dib SA, de Almeida-Pititto B. The gut microbiome of obese postpartum women with and without previous gestational diabetes mellitus and the gut microbiota of their babies. Diabetol Metab Syndr 2022; 14:194. [PMID: 36566315 PMCID: PMC9790115 DOI: 10.1186/s13098-022-00954-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 11/14/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The incidence of gestational diabetes mellitus (GDM) is increasing worldwide, and has been associated with some changes in the gut microbiota. Studies have shown that the maternal gut microbiota pattern with hyperglycemia can be transmitted to the offspring. The study aimed to evaluate the gut microbiota of obese postpartum women with and without previous GDM and their offspring. METHODS We evaluated a total of 84 puerperal women who had (n = 40) or not GDM (n = 44), and their infants were also included. Stool samples were obtained 2-6 months after delivery. The molecular profile of the fecal microbiota was obtained by sequencing V4 region of 16S rRNA gene (Illumina® MiSeq). RESULTS We found that the gut microbiota structures of the puerperal women and their infants were similar. Stratifying according to the type of delivery, the relative abundance of Victivallis genus was higher in women who had natural delivery. Exposure to exclusive breastfeeding was associated with a greater abundance of Bacteroides and Staphylococcus. The differential abundance test showed correlations to clinical and laboratory parameters. This work showed no difference in the microbiota of obese puerperal women with and without GDM and their offspring. However, breastfeeding contributed to the ecological succession of the intestinal microbiota of the offspring. CONCLUSION This work can contribute to understanding the potential effects of GDM and early life events on the gut microbiome of mothers and their offspring and its possible role in metabolism later in life.
Collapse
Affiliation(s)
- Patricia Medici Dualib
- Department of Medicine, Escola Paulista Medicina, Universidade Federal de São Paulo, Rua Sena Madureira, 1500, Vila Clementino, São Paulo, SP, CEP 04021-001, Brazil.
| | - Gabriel Fernandes
- DepaBiosystems Informatics and Genomics Group, Instituto René Rachou - Fiocruz Minas, Av Augusto de Lima, 1714, Belo Horizonte, MG, CEP 30190-002, Brazil
| | - Carla R Taddei
- Department of Clinical and Toxicological Analysis, Universidade de São Paulo (USP), Av. Prof. Lineu Prestes 580-Bloco 17, São Paulo, SP, CEP 05508-000, Brazil
| | - Camila R S Carvalho
- Graduate Program in Endocrinology and Metabology, Universidade Federal de São Paulo, Rua Estado de Israel, nº 639, Vila Clementino, São Paulo, SP, CEP 04022-001, Brazil
| | - Luiz Gustavo Sparvoli
- Department of Clinical and Toxicological Analysis, Universidade de São Paulo (USP), Av. Prof. Lineu Prestes 580-Bloco 17, São Paulo, SP, CEP 05508-000, Brazil
| | - Célia Bittencourt
- Graduate Program in Endocrinology and Metabology, Universidade Federal de São Paulo, Rua Estado de Israel, nº 639, Vila Clementino, São Paulo, SP, CEP 04022-001, Brazil
| | - Isis T Silva
- Nutrition Course, Centro Universitário Estácio de Sá, Rua Erê, 207, Belo Horizonte, MG, CEP 30411-052, Brazil
| | - Rosiane Mattar
- Departament of Obstetrics, Escola Paulista de Medicina, Universidade Federal de São Paulo, R. Napoleão de Barros, 875 - Vila Clementino, São Paulo, SP, CEP 04024-002, Brazil
| | - Sandra R G Ferreira
- Department of Epidemiology, School of Public Health, Universidade de São Paulo, Av. Dr. Arnaldo, 715 - Cerqueira César, São Paulo, SP, CEP 01246-904, Brazil
| | - Sergio A Dib
- Department of Medicine, Escola Paulista Medicina, Universidade Federal de São Paulo, Rua Sena Madureira, 1500, Vila Clementino, São Paulo, SP, CEP 04021-001, Brazil
| | - Bianca de Almeida-Pititto
- Department of Preventive Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, Campus São Paulo, Rua Botucatu, n° 740, Vila Clementino, São Paulo, SP, CEP 04023-062, Brazil
| |
Collapse
|
104
|
Lin H, He S, Tan B, Zhang X, Lin Y, Yang Q. Effect of Rice Protein Meal Replacement of Fish Meal on Growth, Anti-Oxidation Capacity, and Non-Specific Immunity for Juvenile Shrimp Litopenaeus vannamei. Animals (Basel) 2022; 12:3579. [PMID: 36552499 PMCID: PMC9774461 DOI: 10.3390/ani12243579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
This study assessed the effect of rice protein meal replacement for fish meal on the growth, nonspecific immunity, and disease resistance on juvenile shrimp Litopenaeus vannamei. Six groups of iso-nitrogenous and iso-lipid feeds named FM, R10, R20, R40, R60, and R80 were prepared by replacing 0%, 10%, 20%, 40%, 60%, and 80% in FM protein with RPM, respectively, and then fed to the shrimps (0.54 ± 0.01 g). An amount of 720 healthy and evenly sized shrimps were allocated to six groups (three replicates per group) and fed four times a day (7:00, 11:00, 17:00 and 21:00) for eight weeks. Results revealed no significant differences in WG, FCR, and SGR of shrimps after replacing FM with 10% RPM (p > 0.05). In the R10 and R20 groups, SOD and T-AOC activities were significantly higher than those in the FM group, whereas the opposite was observed for MDA content (p < 0.05). CAT, ACP, and LZM were all significantly higher in the R10, R20, and R40 groups than in the FM group (p < 0.05). GSH-Px activity in the R10 group was significantly higher than the activity in the FM group (p < 0.05). AKP, PO, TYS, GPT, and GOT activities were significantly higher in the R10 group than in the FM group (p < 0.05). Compared to the FM group, the eukaryotic translation initiation factor 3K (eif3k) gene was significantly up-regulated in the R10 group, whereas the penaiedin 3a (pen 3a) and anti-lipopolysaccharide factor (alf) genes were significantly up-regulated in the R10 and R20 groups (p < 0.05). The crustin a (cru a), immune deficiency (imd), and lysozyme (lzm) mRNA levels were significantly higher in the R10, R20, and R40 groups than in the other groups (p < 0.05). The prophenoloxidase (PO) mRNA levels in the R20 group were significantly higher than those in the FM group (p < 0.05). The replacement of 10−40% of FM with RPM improved the gut flora composition of shrimps, increasing beneficial bacteria (Bacteroidetes) abundance and reducing harmful bacteria (Aspergillus and Vibrio) abundance. After the challenge test of Vibrio parahaemolyticus (7 days), the cumulative mortality in the R10 group significantly decreased (p < 0.05). In conclusion, replacement of 10% FM by RPM significantly improved digestibility, protein synthesis, antioxidant capacity, and disease resistance in L. vannamei.
Collapse
Affiliation(s)
- Huaxing Lin
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang 524088, China
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| | - Shuqing He
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang 524088, China
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| | - Beiping Tan
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang 524088, China
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| | - Xiaomin Zhang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang 524088, China
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| | - Yi Lin
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang 524088, China
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| | - Qihui Yang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang 524088, China
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| |
Collapse
|
105
|
Yilmaz B, Fuhrer T, Morgenthaler D, Krupka N, Wang D, Spari D, Candinas D, Misselwitz B, Beldi G, Sauer U, Macpherson AJ. Plasticity of the adult human small intestinal stoma microbiota. Cell Host Microbe 2022; 30:1773-1787.e6. [PMID: 36318918 DOI: 10.1016/j.chom.2022.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/18/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022]
Abstract
The human distal small intestine (ileum) has a distinct microbiota, but human studies investigating its composition and function have been limited by the inaccessibility of the ileum without purging and/or deep intubation. We investigated inherent instability, temporal dynamics, and the contribution of fed and fasted states using stoma samples from cured colorectal cancer patients as a non-invasive access route to the otherwise inaccessible small and large intestines. Sequential sampling of the ileum before and after stoma formation indicated that ileostoma microbiotas represented that of the intact small intestine. Ileal and colonic stoma microbiotas were confirmed as distinct, and two types of instability in ileal host-microbial relationships were observed: inter-digestive purging followed by the rapid postprandial blooming of bacterial biomass and sub-strain appearance and disappearance within individual taxa after feeding. In contrast to the relative stability of colonic microbiota, the human small intestinal microbiota biomass and its sub-strain composition can be highly dynamic.
Collapse
Affiliation(s)
- Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland; Bern Center for Precision Medicine (BCPM), University of Bern, 3008 Bern, Switzerland.
| | - Tobias Fuhrer
- Institute of Molecular Systems Biology, Swiss Federal Institute of Technology (ETH) Zürich, 8093 Zürich, Switzerland
| | - Deborah Morgenthaler
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland
| | - Niklas Krupka
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland
| | - Daoming Wang
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands; Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands
| | - Daniel Spari
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland
| | - Daniel Candinas
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland
| | - Benjamin Misselwitz
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland
| | - Guido Beldi
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland
| | - Uwe Sauer
- Institute of Molecular Systems Biology, Swiss Federal Institute of Technology (ETH) Zürich, 8093 Zürich, Switzerland
| | - Andrew J Macpherson
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland; Bern Center for Precision Medicine (BCPM), University of Bern, 3008 Bern, Switzerland.
| |
Collapse
|
106
|
Dania MI, Faraji B, Wachira J. Micronutrient Biosynthesis Potential of Spontaneous Grain Fermentation Microbiomes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:16621. [PMID: 36554499 PMCID: PMC9778892 DOI: 10.3390/ijerph192416621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/28/2022] [Accepted: 12/03/2022] [Indexed: 06/17/2023]
Abstract
Fermented foods play an important role in the human diet and particularly so in under-resourced environments where cold preservation is not attainable due to irregular supply of electricity. Fermented foods are reported to support gut health by contributing probiotics. The purpose of this study was to investigate the microbial diversity and metabolic potential of spontaneous millet fermentation. The literature in the field was reviewed and analyses were conducted on publicly available Sequence Read Archive (SRA) datasets. Quality analysis was performed with FastQC, and operational taxonomic units (OTUs) were generated using Quantitative Insights Into Microbial Ecology (QIIME2) and Divisive Amplicon Denoising Algorithm (DADA2) pipelines with Greengenes as the reference database. Metagenomics and pathways analysis were performed with Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt2). Statistical analysis and visualization were accomplished with Statistical Analysis of Metagenomic Profiles (STAMP). At the family taxonomic level, there were differences in the relative abundances of the dominant taxa of bacteria that are involved in the spontaneous fermentation of millet namely Lactobacillaceae, Burkholderiaceae, Streptococcaceae, Leuconostocaceae, and Acetobacteraceae. Clostridiaceae was the dominant family in one dataset. The incidence of Lactobacillaceae and Bifidobacteriaceae suggest the probiotic characteristics of fermented millet. The datasets were collected with fermentations that were mediated by autochthonous microorganisms and the presence of some potential pathogens such as Enterobacteriaceae, Clostridiaceae, Aeromonadaceae, Microbacteiaceae, Pseudomonadaceae, and Neisseriaceae which suggest the need for standardization of fermentation approaches. The genomes show the potential to synthesize metabolites such as essential amino acids and vitamins, suggesting that the respective fermented foods can be further optimized to enhance nutritional benefits.
Collapse
Affiliation(s)
- Margaret I. Dania
- Food Technology Department, Auchi Polytechnic, Auchi 312001, Nigeria
| | - Bahram Faraji
- Nutritional Science/Dietetics Program, Morgan State University, Baltimore, MD 21251, USA
| | - James Wachira
- Department of Biology, Morgan State University, Baltimore, MD 21251, USA
| |
Collapse
|
107
|
Chen Z, Sun Y, Chen L, Zhang Y, Wang J, Li H, Yan X, Xia L, Yao G. Differences in meat quality between Angus cattle and Xinjiang brown cattle in association with gut microbiota and its lipid metabolism. Front Microbiol 2022; 13:988984. [PMID: 36560955 PMCID: PMC9763702 DOI: 10.3389/fmicb.2022.988984] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota plays important roles in mediating fat metabolic events in humans and animals. However, the differences of meat quality traits related to the lipid metabolism (MQT-LM) in association with gut microbiota involving in lipid metabolism have not been well explored between Angus cattle (AG) and Xinjiang brown cattle (BC). Ten heads of 18-month-old uncastrated male AG and BC (5 in each group) raised under the identical conditions were selected to test MQT-LM, i.e., the backfat thickness (BFT), the intramuscular fat (IMF) content, the intramuscular adipocyte areas (IAA), the eye muscle area (EMA), the muscle fiber sectional area (MFSA) and the muscle shear force after sacrifice. The gut microbiota composition and structure with its metabolic function were analyzed by means of metagenomics and metabolomics with rectal feces. The correlation of MQT-LM with the gut microbiota and its metabolites was analyzed. In comparison with AG, BC had significant lower EMA, IMF content and IAA but higher BFT and MFSA. Chao1 and ACE indexes of α-diversity were lower. β-diversity between AG and BC were significantly different. The relative abundance of Bacteroidetes, Prevotella and Blautia and Prevotella copri, Blautia wexlerae, and Ruminococcus gnavus was lower. The lipid metabolism related metabolites, i.e., succinate, oxoglutaric acid, L-aspartic acid and L-glutamic acid were lower, while GABA, L-asparagine and fumaric acid were higher. IMF was positively correlated with Prevotella copri, Blautia wexlerae and Ruminococcus gnavus, and the metabolites succinate, oxoglutaric acid, L-aspartic acid and L-glutamic acid, while negatively with GABA, L-asparagine and fumaric acid. BFT was negatively correlated with Blautia wexlerae and the metabolites succinate, L-aspartic acid and L-glutamic acid, while positively with GABA, L-asparagine and fumaric acid. Prevotella Copri, Blautia wexlerae, and Ruminococcus gnavus was all positively correlated with succinate, oxoglutaric acid, while negatively with L-asparagine and fumaric acid. In conclusion, Prevotella copri, Prevotella intermedia, Blautia wexlerae, and Ruminococcus gnavus may serve as the potential differentiated bacterial species in association with MQT-LM via their metabolites of oxoglutaric acid, succinate, fumaric acid, L-aspartic acid, L-asparagine, L-glutamic acid and GABA between BC and AG.
Collapse
Affiliation(s)
- Zhuo Chen
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Yawei Sun
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Lijing Chen
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Yang Zhang
- Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Jinquan Wang
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Hongbo Li
- Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Xiangming Yan
- Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Lining Xia
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China,Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJKLNDSCHA), Xinjiang Agricultural University, Urumqi, China,*Correspondence: Lining Xia,
| | - Gang Yao
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China,Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJKLNDSCHA), Xinjiang Agricultural University, Urumqi, China,Gang Yao,
| |
Collapse
|
108
|
Gao Z, Song H, Dong H, Ji X, Lei Z, Tian Y, Wu Y, Zou H. Comparative analysis of intestinal flora between rare wild red-crowned crane and white-naped crane. Front Microbiol 2022; 13:1007884. [PMID: 36532425 PMCID: PMC9752901 DOI: 10.3389/fmicb.2022.1007884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 11/14/2022] [Indexed: 10/17/2024] Open
Abstract
INTRODUCTION Animal intestines are extremely rich in microbial ecosystems. Numerous studies in different fields, such as epidemiology and histology, have revealed that gut microorganisms considerably mediate the survival and reproduction of animals. However, gut microbiology studies of homogeneously distributed wild cranes are still rare. This study aimed to understand the structural composition of the gut microbial community of wild cranes and elucidate the potential roles of the microorganisms. METHODS We used high-throughput sequencing to analyze the gut microbial community structure of wild cranes in the Zhalong Nature Reserve. RESULTS A total of 1,965,683 valid tags and 5248 OTUs were obtained from 32 fecal samples. Twenty-six bacteria phyla and 523 genera were annotated from the intestinal tract of the red-crowned crane. Twenty-five bacteria phyla and 625 genera were annotated from the intestine of the white-naped crane. Firmicutes, Proteobacteria, and Bacteroidetes are the dominant bacterial phyla in the intestinal tract of red-crowned cranes, while Catellicoccus, Lactobacillus, Neisseria, and Streptococcus were the dominant genera. The dominant bacterial phyla in the intestinal tract of white-naped cranes were Firmicutes, Proteobacteria, Bacteroidetes, Epsilonbacteraeota, Actinobacteria, and Fusobacteria. However, the dominant genera were Catellicoccus, Lactobacillus, Neisseria, Campylobacter, Streptococcus, Anaerobiospirillum, Romboutsia, Turicibacter, Haemophilus, and Lautropia. Firmicutes had significantly higher relative abundance in the intestine of the red-crowned than white-naped cranes (P < 0.05). However, the relative abundance of Actinobacteria and Bacteroidetes was significantly higher (P < 0.05) in the intestines of white-naped than red-crowned cranes. The diversity of the intestinal flora between the two crane species was significantly different (P < 0.05). Besides, the alpha diversity of the intestinal flora was higher for white-naped than red-crowned cranes. Eight of the 41 functional pathways differed in the gut of both crane species (P < 0.05). DISCUSSION Both species live in the same area and have similar feeding and behavioral characteristics. Therefore, host differences are possibly the main factors influencing the structural and functional differences in the composition of the gut microbial community. This study provides important reference data for constructing a crane gut microbial assessment system. The findings have implications for studying deeper relationships between crane gut microbes and genetics, nutrition, immunity, and disease.
Collapse
Affiliation(s)
- Zhongsi Gao
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| | - Hongwei Song
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| | - Haiyan Dong
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| | - Xiaolong Ji
- Department of Genetics, College of Life Science, Northeast Forestry University, Harbin, China
| | - Zefeng Lei
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| | - Ye Tian
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| | - Yining Wu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| | - Hongfei Zou
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| |
Collapse
|
109
|
Chai L, Jabbie IS, Chen A, Jiang L, Li M, Rao H. Effects of waterborne Pb/Cu mixture on Chinese toad, Bufo gargarizans tadpoles: morphological, histological, and intestinal microbiota assessment. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:90656-90670. [PMID: 35871197 DOI: 10.1007/s11356-022-22143-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/18/2022] [Indexed: 06/15/2023]
Abstract
Coexistence of heavy metals in aquatic environments exert complex effects on amphibians. Here, the adverse effects of Pb (0.14 μM) combined with Cu at concentrations of 0, 0.25, and 1.0 μM were investigated in Bufo gargarizans tadpoles. Tadpoles were chronically exposed from Gosner stage (Gs) 26 to Gs 38, and morphology of tadpoles as well as intestinal histology and bacterial community were assessed. Our results indicated that Pb+Cu1.0 exposure induced significant retardation of somatic mass, total length, intestine mass, and intestine length as well as intestinal histological alterations. Pb+Cu0.25 and Pb+Cu1.0 exposure were associated with the loss of gut bacterial diversity. Proteobacteria and Bacteroidetes were two dominant phyla in tadpoles independently of heavy metal exposure, but the abundance of Proteobacteria increased significantly in Pb+Cu1.0 group and Bacteroidetes decreased significantly in all treatment groups. Furthermore, functional prediction indicated that metabolic disorders were associated with Pb+Cu0.25 and Pb+Cu1.0 exposure. Overall, relative limited shifts in intestinal bacterial diversity, composition, and functionality caused by Pb+Cu0 exposure, while coexistence of Pb and Cu induced gut dysbiosis and might further cause disturbance of metabolic homeostasis. The findings of this study provide insights into the effects of Pb and Cu coexistence on the health of amphibians.
Collapse
Affiliation(s)
- Lihong Chai
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China.
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, China.
| | - Ibrahim Sory Jabbie
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, China
| | - Aixia Chen
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, China
| | - Ling Jiang
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, China
| | - Mengfan Li
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, China
| | - Huihui Rao
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, China
| |
Collapse
|
110
|
Effect of diet on gut microbiota diversity in mandarin ducks (Aix galericulata) revealed by Illumina high-throughput sequencing. Arch Microbiol 2022; 204:725. [DOI: 10.1007/s00203-022-03333-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022]
|
111
|
Food for the mind: The journey of probiotics from foods to ANTI-Alzheimer’s disease therapeutics. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
112
|
Li Z, Liu R, Wang X, Wu H, Yi X, Huang L, Qin Q. Effects of melittin on laying performance and intestinal barrier function of quails. Poult Sci 2022; 102:102355. [PMID: 36502563 PMCID: PMC9763859 DOI: 10.1016/j.psj.2022.102355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022] Open
Abstract
To study the effects of melittin on egg-laying performance and intestinal barrier of quails, 240 quails (aged 70 d) were randomly divided into 4 groups with 6 replicates (10 quails per replicate). They were fed with basal diet (group B), basal diet + 0.08 g/kg melittin (group BA1), basal diet + 0.12 g/kg melittin (group BA2) and basal diet + 0.16 g/kg melittin (group BA3). The experiment lasted for 21 days. The eggs were collected every day. At the end of the experiment, duodenal, jejunal, and ileal tissues were collected, and the cecal contents were sampled. Intestinal antioxidant index, barrier function, and intestinal flora were analyzed. The results showed that the addition of melittin significantly increased the laying rate and average egg weight. Addition of melittin significantly increased the antioxidant function, mechanical barrier, immune barrier, and the villus height to crypt depth ratio of small intestine. Addition of melittin had no significant effect on the α and β diversity of cecal flora, but significantly increased the abundance of Bacteroidales at family level and genus level. Bioinformatics analysis of cecal content showed significant increase in COG functional category of cytoskeleton, and significant decrease in RNA processing and modification in group BA2. KEGG functional analysis showed significant decrease in steroid biosynthesis, caffeine metabolism, and cytochrome P450 pathways in group BA2. In conclusion, addition of 0.12 g/kg melittin to feed improved the laying performance and the intestinal antioxidant capacity and barrier function of quails but had no significant effect on the composition and structure of cecal microbial community. This study provides experimental data and theoretical basis for the application of melittin as a new quail feed additive.
Collapse
|
113
|
Zhou C, Li J, Guo C, Zhou Z, Yang Z, Zhang Y, Jiang J, Cai Y, Zhou J, Ming Y. Comparison of intestinal flora between patients with chronic and advanced Schistosoma japonicum infection. Parasit Vectors 2022; 15:413. [PMID: 36345042 PMCID: PMC9640844 DOI: 10.1186/s13071-022-05539-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022] Open
Abstract
Background Schistosoma japonicum infection is an important public health problem, imposing heavy social and economic burdens in 78 countries worldwide. However, the mechanism of transition from chronic to advanced S. japonicum infection remains largely unknown. Evidences suggested that gut microbiota plays a role in the pathogenesis of S. japonicum infection. However, the composition of the gut microbiota in patients with chronic and advanced S. japonicum infection is not well defined. In this study, we compared the composition of the intestinal flora in patients with chronic and advanced S. japonicum infection. Methods The feces of 24 patients with chronic S. japonicum infection and five patients with advanced S. japonicum infection from the same area were collected according to standard procedures, and 16S rRNA sequencing technology was used to analyze the intestinal microbial composition of the two groups of patients. Results We found that alteration occurs in the gut microbiota between the groups of patients with chronic and advanced S. japonicum infections. Analysis of alpha and beta diversity indicated that the diversity and abundance of intestinal flora in patients with advanced S. japonicum infection were lower than those in patients with chronic S. japonicum infection. Furthermore, Prevotella 9, Subdoligranulum, Ruminococcus torques, Megamonas and Fusicatenibacter seemed to have potential to discriminate different stages of S. japonicum infection and to act as biomarkers for diagnosis. Function prediction analysis revealed that microbiota function in the chronic group was focused on translation and cell growth and death, while that in the advanced group was concentrated on elevating metabolism-related functions. Conclusions Our study demonstrated that alteration in gut microbiota in different stages of S. japonicum infection plays a potential role in the pathogenesis of transition from chronic to advanced S. japonicum infection. However, further validation in the clinic is needed, and the underlying mechanism requires further study. Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05539-6.
Collapse
|
114
|
Jiang LY, Kan YN, Yu ZP, Jian BY, Yao SJ, Lv LY, Liu JC. Prebiotic Effects of Chinese Herbal Polysaccharides on NAFLD Amelioration: The Preclinical Progress. Nat Prod Commun 2022; 17. [DOI: 10.1177/1934578x221124751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is caused by fatty degeneration of liver cells, and there are currently no effective treatments. Numerous investigations have demonstrated that Chinese herbal medicines (CHMs) are effective against NAFLD. Polysaccharides (PS), the major components of most CHM, are primarily taken orally to be degraded and fermented by gut microbiota, which makes them a promising multivalent and multifunctional prebiotic candidate for NAFLD. In this review, the experimental evidence to prevent and treat NAFLD using the unique prebiotic effects of PS isolated from CHM are summarized to discuss additional treatment options for NAFLD.
Collapse
Affiliation(s)
- Li-Yan Jiang
- Department of Pathogen Biology, Medical Technology College of Qiqihar Medical University, Qiqihar, China
| | - Yu-Na Kan
- Department of Polygenic Diseases, Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Zhi-Pu Yu
- Department of Equipment, The Second Affiliated Hospital, Qiqihar Medical University, Qiqihar, China
| | - Bai-Yu Jian
- Department of Polygenic Diseases, Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Shu-Juan Yao
- Department of Pathogen Biology, Medical Technology College of Qiqihar Medical University, Qiqihar, China
| | - Li-Yan Lv
- Department of Pathogen Biology, Medical Technology College of Qiqihar Medical University, Qiqihar, China
| | - Ji-Cheng Liu
- Department of Polygenic Diseases, Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
115
|
Chompre G, Sambolin L, Cruz ML, Sanchez R, Rodriguez Y, Rodríguez-Santiago RE, Yamamura Y, Appleyard CB. A one month high fat diet disrupts the gut microbiome and integrity of the colon inducing adiposity and behavioral despair in male Sprague Dawley rats. Heliyon 2022; 8:e11194. [PMID: 36387539 PMCID: PMC9663868 DOI: 10.1016/j.heliyon.2022.e11194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/17/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
High-fat diet (HFD) is associated with gut microbiome dysfunction and mental disorders. However, the time-dependence as to when this occurs is unclear. We hypothesized that a short-term HFD causes colonic tissue integrity changes resulting in behavioral changes. Rats were fed HFD or low-fat diet (LFD) for a month and gut microbiome, colon, and behavior were evaluated. Behavioral despair was found in the HFD group. Although obesity was absent, the HFD group showed increased percent weight gain, epididymal fat tissue, and leptin expression. Moreover, the HFD group had increased colonic damage, decreased expression of the tight junction proteins, and higher lipopolysaccharides (LPS) in serum. Metagenomic analysis revealed that the HFD group had more Bacteroides and less S24-7 which correlated with the decreased claudin-5. Finally, HFD group showed an increase of microglia percent area, increased astrocytic projections, and decreased phospho-mTOR. In conclusion, HFD consumption in a short period is still sufficient to disrupt gut integrity resulting in LPS infiltration, alterations in the brain, and behavioral despair even in the absence of obesity.
Collapse
Affiliation(s)
- Gladys Chompre
- Biology and Biotechnology Department, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico
- Basic Sciences Department, Division of Physiology, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Lubriel Sambolin
- Basic Sciences Department, Division of Pharmacology, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Myrella L. Cruz
- Basic Sciences Department, Division of Physiology, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Rafael Sanchez
- AIDS Research Infrastructure Program, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Yarelis Rodriguez
- Basic Sciences Department, Division of Physiology, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Ronald E. Rodríguez-Santiago
- AIDS Research Infrastructure Program, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Yasuhiro Yamamura
- AIDS Research Infrastructure Program, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Caroline B. Appleyard
- Basic Sciences Department, Division of Physiology, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| |
Collapse
|
116
|
Mousa WK, Chehadeh F, Husband S. Microbial dysbiosis in the gut drives systemic autoimmune diseases. Front Immunol 2022; 13:906258. [PMID: 36341463 PMCID: PMC9632986 DOI: 10.3389/fimmu.2022.906258] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/20/2022] [Indexed: 09/29/2023] Open
Abstract
Trillions of microbes survive and thrive inside the human body. These tiny creatures are crucial to the development and maturation of our immune system and to maintain gut immune homeostasis. Microbial dysbiosis is the main driver of local inflammatory and autoimmune diseases such as colitis and inflammatory bowel diseases. Dysbiosis in the gut can also drive systemic autoimmune diseases such as type 1 diabetes, rheumatic arthritis, and multiple sclerosis. Gut microbes directly interact with the immune system by multiple mechanisms including modulation of the host microRNAs affecting gene expression at the post-transcriptional level or production of microbial metabolites that interact with cellular receptors such as TLRs and GPCRs. This interaction modulates crucial immune functions such as differentiation of lymphocytes, production of interleukins, or controlling the leakage of inflammatory molecules from the gut to the systemic circulation. In this review, we compile and analyze data to gain insights into the underpinning mechanisms mediating systemic autoimmune diseases. Understanding how gut microbes can trigger or protect from systemic autoimmune diseases is crucial to (1) tackle these diseases through diet or lifestyle modification, (2) develop new microbiome-based therapeutics such as prebiotics or probiotics, (3) identify diagnostic biomarkers to predict disease risk, and (4) observe and intervene with microbial population change with the flare-up of autoimmune responses. Considering the microbiome signature as a crucial player in systemic autoimmune diseases might hold a promise to turn these untreatable diseases into manageable or preventable ones.
Collapse
Affiliation(s)
- Walaa K. Mousa
- Biology Department, Whitman College, Walla Walla, WA, United States
- College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates
- College of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Fadia Chehadeh
- Biology Department, Whitman College, Walla Walla, WA, United States
| | - Shannon Husband
- Biology Department, Whitman College, Walla Walla, WA, United States
| |
Collapse
|
117
|
Gibiino G, Binda C, Cristofaro L, Sbrancia M, Coluccio C, Petraroli C, Jung CFM, Cucchetti A, Cavaliere D, Ercolani G, Sambri V, Fabbri C. Dysbiosis and Gastrointestinal Surgery: Current Insights and Future Research. Biomedicines 2022; 10:2532. [PMID: 36289792 PMCID: PMC9599064 DOI: 10.3390/biomedicines10102532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
Surgery of the gastrointestinal tract can result in deep changes among the gut commensals in terms of abundance, function and health consequences. Elective colorectal surgery can occur for neoplastic or inflammatory bowel disease; in these settings, microbiota imbalance is described as a preoperative condition, and it is linked to post-operative complications, as well. The study of bariatric patients led to several insights into the role of gut microbiota in obesity and after major surgical injuries. Preoperative dysbiosis and post-surgical microbiota reassessment are still poorly understood, and they could become a key part of preventing post-surgical complications. In the current review, we outline the most recent literature regarding agents and molecular pathways involved in pre- and post-operative dysbiosis in patients undergoing gastrointestinal surgery. Defining the standard method for microbiota assessment in these patients could set up the future approach and clinical practice.
Collapse
Affiliation(s)
- Giulia Gibiino
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
| | - Cecilia Binda
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
| | - Ludovica Cristofaro
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
- Department of Medical and Surgical Sciences—DIMEC, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy
| | - Monica Sbrancia
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
| | - Chiara Coluccio
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
| | - Chiara Petraroli
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
| | - Carlo Felix Maria Jung
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
| | - Alessandro Cucchetti
- Department of Medical and Surgical Sciences—DIMEC, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy
- General and Oncologic Surgery, Morgagni—Pierantoni Hospital, AUSL Romagna, 47121 Forlì-Cesena, Italy
| | - Davide Cavaliere
- General and Oncologic Surgery, Morgagni—Pierantoni Hospital, AUSL Romagna, 47121 Forlì-Cesena, Italy
| | - Giorgio Ercolani
- Department of Medical and Surgical Sciences—DIMEC, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy
- General and Oncologic Surgery, Morgagni—Pierantoni Hospital, AUSL Romagna, 47121 Forlì-Cesena, Italy
| | - Vittorio Sambri
- Department of Medical and Surgical Sciences—DIMEC, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy
- Microbiology Unit, Hub Laboratory, AUSL della Romagna, 47121 Forlì-Cesena, Italy
| | - Carlo Fabbri
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
| |
Collapse
|
118
|
Raj K, Singh S, Chib S, Mallan S. Microbiota- Brain-Gut-Axis Relevance to Parkinson's Disease: Potential Therapeutic Effects of Probiotics. Curr Pharm Des 2022; 28:3049-3067. [PMID: 36200207 DOI: 10.2174/1381612828666221003112300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/02/2022] [Indexed: 01/28/2023]
Abstract
Parkinson's disease (PD) is the second most common type of neurogenerative disease among middleaged and older people, characterized by aggregation of alpha-synuclein and dopaminergic neuron loss. The microbiota- gut-brain axis is a dynamic bidirectional communication network and is involved in the pathogenesis of PD. The aggregation of misfolded protein alpha-synuclein is a neuropathological characteristic of PD, originates in the gut and migrates to the central nervous system (CNS) through the vagus nerve and olfactory bulb. The change in the architecture of gut microbiota increases the level short-chain fatty acids (SCFAs) and other metabolites, acting on the neuroendocrine system and modulating the concentrations of gamma-Aminobutyric acid (GABA), serotonin, and other neurotransmitters. It also alters the vagus and intestinal signalling, influencing the brain and behaviour by activating microglia and systemic cytokines. Both experimental and clinical reports indicate the role of intestinal dysbiosis and microbiota host interaction in neurodegeneration. Probiotics are live microorganisms that modify the gut microbiota in the small intestine to avoid neurological diseases. Probiotics have been shown in clinical and preclinical studies to be effective in the treatment of PD by balancing the gut microbiota. In this article, we described the role of gut-microbiota in the pathogenesis of PD. The article aims to explore the mechanistic strategy of the gut-brain axis and its relation with motor impairment and the use of probiotics to maintain gut microbial flora and prevent PD-like symptoms.
Collapse
Affiliation(s)
- Khadga Raj
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab 142001, India
| | - Shamsher Singh
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab 142001, India
| | - Shivani Chib
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab 142001, India
| | - Sudhanshu Mallan
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab 142001, India
| |
Collapse
|
119
|
Gómez-Duarte OG, Ogra PL. Editorial: Impact of microbiome on gut mucosal immunity in health and disease. Front Immunol 2022; 13:1005156. [PMID: 36211406 PMCID: PMC9532250 DOI: 10.3389/fimmu.2022.1005156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/07/2022] [Indexed: 12/02/2022] Open
Affiliation(s)
| | - Pearay L. Ogra
- *Correspondence: Oscar G. Gómez-Duarte, ; Pearay L. Ogra,
| |
Collapse
|
120
|
Wu Z, Shen J, Xu Q, Xiang Q, Chen Y, Lv L, Zheng B, Wang Q, Wang S, Li L. Epigallocatechin-3-Gallate Improves Intestinal Gut Microbiota Homeostasis and Ameliorates Clostridioides difficile Infection. Nutrients 2022; 14:nu14183756. [PMID: 36145133 PMCID: PMC9504111 DOI: 10.3390/nu14183756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/04/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Clostridioides difficile infection is closely related to the intestinal flora disorders induced by antibiotics, and changes in the intestinal flora may cause the occurrence and development of Clostridioides difficile infection. Epigallocatechin-3-gallate (EGCG) is one of the major bioactive ingredients of green tea and has been suggested to alleviate the growth of C. difficile in vitro. EGCG can ameliorate several diseases, such as obesity, by regulating the gut microbiota. However, whether EGCG can attenuate C. difficile infection by improving the gut microbiota is unknown. After establishing a mouse model of C. difficile infection, mice were administered EGCG (25 or 50 mg/kg/day) or PBS intragastrically for 2 weeks to assess the benefits of EGCG. Colonic pathology, inflammation, the intestinal barrier, gut microbiota composition, metabolomics, and the transcriptome were evaluated in the different groups. Compared with those of the mice in the CDI group, EGCG improved survival rates after infection, improved inflammatory markers, and restored the damage to the intestinal barrier. Furthermore, EGCG could improve the intestinal microbial community caused by C. difficile infection, such as by reducing the relative abundance of Enterococcaceae and Enterobacteriaceae. Moreover, EGCG can increase short-chain fatty acids, improve amino acid metabolism, and downregulate pathways related to intestinal inflammation. EGCG alters the microbiota and alleviates C. difficile infection, which provides new insights into potential therapies.
Collapse
Affiliation(s)
- Zhengjie Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Jian Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Qiaomai Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Qiangqiang Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Yunbo Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Beiwen Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Qiangqiang Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Shuting Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China
- Correspondence: ; Tel.: +86-571-8723-6458; Fax: +86-571-8723-6459
| |
Collapse
|
121
|
Huyan L, Kumar A, Manafi M, Soumeh EA. Effects of protease supplementation on growth performance, organ development, gut morphology, and microbial profile of broiler chicken. ACTA AGR SCAND A-AN 2022. [DOI: 10.1080/09064702.2022.2113121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Lingjun Huyan
- School of Agriculture and Food Sciences, University of Queensland, Gatton, Australia
| | - Arun Kumar
- School of Agriculture and Food Sciences, University of Queensland, Gatton, Australia
| | - Milad Manafi
- School of Agriculture and Food Sciences, University of Queensland, Gatton, Australia
| | - Elham A. Soumeh
- School of Agriculture and Food Sciences, University of Queensland, Gatton, Australia
| |
Collapse
|
122
|
Mhd Omar NA, Dicksved J, Kruger J, Zamaratskaia G, Michaëlsson K, Wolk A, Frank J, Landberg R. Effect of a diet rich in galactose or fructose, with or without fructooligosaccharides, on gut microbiota composition in rats. Front Nutr 2022; 9:922336. [PMID: 36034892 PMCID: PMC9412906 DOI: 10.3389/fnut.2022.922336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Recent studies suggest that a diet rich in sugars significantly affects the gut microbiota. Adverse metabolic effects of sugars may partly be mediated by alterations of gut microbiota and gut health parameters, but experimental evidence is lacking. Therefore, we investigated the effects of high intake of fructose or galactose, with/without fructooligosaccharides (FOS), on gut microbiota composition in rats and explored the association between gut microbiota and low-grade systemic inflammation. Sprague-Dawley rats (n = 6/group) were fed the following isocaloric diets for 12 weeks (% of the dry weight of the sugars or FOS): (1) starch (control), (2) fructose (50%), (3) galactose (50%), (4) starch+FOS (15%) (FOS control), (5) fructose (50%)+FOS (15%), (6) galactose (50%)+FOS (15%), and (7) starch+olive (negative control). Microbiota composition in the large intestinal content was determined by sequencing amplicons from the 16S rRNA gene; 341F and 805R primers were used to generate amplicons from the V3 and V4 regions. Actinobacteria, Verrucomicrobia, Tenericutes, and Cyanobacteria composition differed between diets. Bifidobacterium was significantly higher in all diet groups where FOS was included. Modest associations between gut microbiota and metabolic factors as well as with gut permeability markers were observed, but no associations between gut microbiota and inflammation markers were observed. We found no coherent effect of galactose or fructose on gut microbiota composition. Added FOS increased Bifidobacterium but did not mitigate potential adverse metabolic effects induced by the sugars. However, gut microbiota composition was associated with several metabolic factors and gut permeability markers which warrant further investigations.
Collapse
Affiliation(s)
- Nor Adila Mhd Omar
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johan Dicksved
- Department of Animal Nutrition and Management, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Johanita Kruger
- Department of Food Biofunctionality, Institute of Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
| | - Galia Zamaratskaia
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Karl Michaëlsson
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Alicja Wolk
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jan Frank
- Department of Food Biofunctionality, Institute of Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
| | - Rikard Landberg
- Department of Public Health and Clinical Medicine, Nutritional Research. Umeå University, Umeå, Sweden.,Division of Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
123
|
ROLE OF GUT MICROBIOTA IN DEPRESSION: UNDERSTANDING MOLECULAR PATHWAYS, RECENT RESEARCH, AND FUTURE DIRECTION. Behav Brain Res 2022; 436:114081. [PMID: 36037843 DOI: 10.1016/j.bbr.2022.114081] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/21/2022]
Abstract
Gut microbiota, also known as the "second brain" in humans because of the regulatory role it has on the central nervous system via neuronal, chemical and immune pathways. It has been proven that there exists a bidirectional communication between the gut and the brain. Increasing evidence supports that this crosstalk is linked to the etiology and treatment of depression. Reports suggest that the gut microbiota control the host epigenetic machinery in depression and gut dysbiosis causes negative epigenetic modifications via mechanisms like histone acetylation, DNA methylation and non-coding RNA mediated gene inhibition. The gut microbiome can be a promising approach for the management of depression. The diet and dietary metabolites like kynurenine, tryptophan, and propionic acid also greatly influence the microbiome composition and thereby, the physiological activities. This review gives a bird-eye view on the pathological updates and currently used treatment approaches targeting the gut microbiota in depression.
Collapse
|
124
|
Li J, Qu Z, Liu F, Jing H, Pan Y, Guo S, Ho CL. Diet‐Based Microbiome Modulation: You are What You Eat. PRINCIPLES IN MICROBIOME ENGINEERING 2022:1-46. [DOI: 10.1002/9783527825462.ch1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
125
|
Fan Y, Xu C, Xie L, Wang Y, Zhu S, An J, Li Y, Tian Z, Yan Y, Yu S, Liu H, Jia B, Wang Y, Wang L, Yang L, Bian Y. Abnormal bile acid metabolism is an important feature of gut microbiota and fecal metabolites in patients with slow transit constipation. Front Cell Infect Microbiol 2022; 12:956528. [PMID: 35967856 PMCID: PMC9366892 DOI: 10.3389/fcimb.2022.956528] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/04/2022] [Indexed: 12/12/2022] Open
Abstract
Destructions in the intestinal ecosystem are implicated with changes in slow transit constipation (STC), which is a kind of intractable constipation characterized by colonic motility disorder. In order to deepen the understanding of the structure of the STC gut microbiota and the relationship between the gut microbiota and fecal metabolites, we first used 16S rRNA amplicon sequencing to evaluate the gut microbiota in 30 STC patients and 30 healthy subjects. The α-diversity of the STC group was changed to a certain degree, and the β-diversity was significantly different, which indicated that the composition of the gut microbiota of STC patients was inconsistent with healthy subjects. Among them, Bacteroides, Parabacteroides, Desulfovibrionaceae, and Ruminiclostridium were significantly upregulated, while Subdoligranulum was significantly downregulated. The metabolomics showed that different metabolites between the STC and the control group were involved in the process of bile acids and lipid metabolism, including taurocholate, taurochenodeoxycholate, taurine, deoxycholic acid, cyclohexylsulfamate, cholic acid, chenodeoxycholate, arachidonic acid, and 4-pyridoxic acid. We found that the colon histomorphology of STC patients was significantly disrupted, and TGR5 and FXR were significantly downregulated. The differences in metabolites were related to changes in the abundance of specific bacteria and patients’ intestinal dysfunction. Analysis of the fecal genomics and metabolomics enabled separation of the STC from controls based on random forest model prediction [STC vs. control (14 gut microbiota and metabolite biomarkers)—Sensitivity: 1, Specificity: 0.877]. This study provided a perspective for the diagnosis and intervention of STC related with abnormal bile acid metabolism.
Collapse
Affiliation(s)
- Yadong Fan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chen Xu
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Lulu Xie
- School of Medicine, Nankai University, Tianjin, China
| | - Ying Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shan Zhu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiren An
- The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Zhikui Tian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiqi Yan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuang Yu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haizhao Liu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Beitian Jia
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiyang Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Li Wang
- The Pharmacy Department, Tianjin Second People's Hospital, Tianjin, China
| | - Long Yang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Long Yang, ; Yuhong Bian,
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Long Yang, ; Yuhong Bian,
| |
Collapse
|
126
|
Takeuchi H, Kato Y, Sasaki N, Tanigaki K, Yamaga S, Mita E, Kuboniwa M, Matsusaki M, Amano A. Surface pre-reacted glass-ionomer eluate protects gingival epithelium from penetration by lipopolysaccharides and peptidoglycans via transcription factor EB pathway. PLoS One 2022; 17:e0271192. [PMID: 35895663 PMCID: PMC9328573 DOI: 10.1371/journal.pone.0271192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 06/24/2022] [Indexed: 11/25/2022] Open
Abstract
Surface pre-reacted glass-ionomer (S-PRG) filler, produced by PRG technology for use with various dental materials, is bioactive and known to release ions from a glass-ionomer phase. We previously reported that coxsackievirus and adenovirus receptor (CXADR), a tight junction associated protein, was located in the epithelial barrier of gingival epithelium. In the present study, the tissue protective effects of an S-PRG eluate prepared with S-PRG filler were investigated using a three-dimensional human gingival epithelial tissue model. The results showed that the S-PRG eluate specifically induced CXADR expression at the transcriptional level of messenger RNA as well as the protein level, and also nuclear translocation of transcription factor EB (TFEB) in gingival epithelial cells. Furthermore, shigyakusan, a TFEB inhibitor, canceled induction of the CXADR protein by the S-PRG eluate. Additionally, gingival epithelial permeation by 40-kDa dextran, lipopolysaccharide, and peptidoglycan in the 3D-tissue models was prevented by the eluate, with those effects abrogated by knockdown of CXADR. These findings suggest that S-PRG eluate increases CXADR expression via the TFEB pathway, thus inhibiting penetration of bacterial virulence factors into subepithelial tissues.
Collapse
Affiliation(s)
- Hiroki Takeuchi
- Department of Preventive Dentistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
- * E-mail:
| | - Yuta Kato
- Department of Preventive Dentistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | - Naoko Sasaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Keita Tanigaki
- Department of Preventive Dentistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | - Shunsuke Yamaga
- Department of Preventive Dentistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | - Ena Mita
- Department of Preventive Dentistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | - Masae Kuboniwa
- Department of Preventive Dentistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | - Michiya Matsusaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Atsuo Amano
- Department of Preventive Dentistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
127
|
Buglione M, Ricca E, Petrelli S, Baccigalupi L, Troiano C, Saggese A, Rivieccio E, Fulgione D. Gut microbiota plasticity in insular lizards under reversed island syndrome. Sci Rep 2022; 12:12682. [PMID: 35879521 PMCID: PMC9314426 DOI: 10.1038/s41598-022-16955-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 07/19/2022] [Indexed: 11/28/2022] Open
Abstract
Animals living on small islands are more drastically exposed to environmental changes, such as food or water starvation, and rapid temperature shifts. Facing such conditions, and probably thank to adaptive plasticity mechanisms, some animals display a Reversed Island Syndrome (RIS), a suite of traits, including skin pigmentation, voracity, sexual dimorphism, showed differently from mainland relatives. Here, we analyse a so far poorly explored aspect of RIS: the effect of this on the microbiota composition of host Italian wall lizard (Podarcis siculus), strongly influenced by the animal’s lifestyle, and conditioning the same. We compare mainland and island populations, assessing the difference between their microbial communities and their response under unexpected food, experimentally provided. Our observations showed a significant difference in microbiota communities between island and mainland groups, depended mainly from changes in relative abundance of the shared genera (difference due to decrease/increase). Exposure to experimental diet regimes resulted into significative reshaping of bacterial composition of microbiota and a greater variation in body mass only in the island population. Our results could be an evidence that gut microbial community contributes to adaptive plasticity mechanisms of island lizards under RIS to efficiently respond to unexpected changes.
Collapse
Affiliation(s)
- Maria Buglione
- Department of Biology, University of Naples Federico II, Via Cinthia 26, 80126, Naples, Italy
| | - Ezio Ricca
- Department of Biology, University of Naples Federico II, Via Cinthia 26, 80126, Naples, Italy.,Task Force of the Federico II University for Microbiome Studies, Naples, Italy
| | - Simona Petrelli
- Department of Biology, University of Naples Federico II, Via Cinthia 26, 80126, Naples, Italy
| | - Loredana Baccigalupi
- Task Force of the Federico II University for Microbiome Studies, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Claudia Troiano
- Department of Biology, University of Naples Federico II, Via Cinthia 26, 80126, Naples, Italy
| | - Anella Saggese
- Department of Biology, University of Naples Federico II, Via Cinthia 26, 80126, Naples, Italy
| | - Eleonora Rivieccio
- Department of Humanities, University of Naples Federico II, Naples, Italy
| | - Domenico Fulgione
- Department of Biology, University of Naples Federico II, Via Cinthia 26, 80126, Naples, Italy. .,Task Force of the Federico II University for Microbiome Studies, Naples, Italy.
| |
Collapse
|
128
|
Jung JH, Kim SE, Suk KT, Kim DJ. Gut microbiota-modulating agents in alcoholic liver disease: Links between host metabolism and gut microbiota. Front Med (Lausanne) 2022; 9:913842. [PMID: 35935787 PMCID: PMC9354621 DOI: 10.3389/fmed.2022.913842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Alcoholic liver disease (ALD) involves a wide spectrum of diseases, including asymptomatic hepatic steatosis, alcoholic hepatitis, hepatic fibrosis, and cirrhosis, which leads to morbidity and mortality and is responsible for 0.9% of global deaths. Alcohol consumption induces bacterial translocation and alteration of the gut microbiota composition. These changes in gut microbiota aggravate hepatic inflammation and fibrosis. Alteration of the gut microbiota leads to a weakened gut barrier and changes host immunity and metabolic function, especially related to bile acid metabolism. Modulation and treatment for the gut microbiota in ALD has been studied using probiotics, prebiotics, synbiotics, and fecal microbial transplantation with meaningful results. In this review, we focused on the interaction between alcohol and gut dysbiosis in ALD. Additionally, treatment approaches for gut dysbiosis, such as abstinence, diet, pro-, pre-, and synbiotics, antibiotics, and fecal microbial transplantation, are covered here under ALD. However, further research through human clinical trials is warranted to evaluate the appropriate gut microbiota-modulating agents for each condition related to ALD.
Collapse
Affiliation(s)
- Jang Han Jung
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, South Korea
| | - Sung-Eun Kim
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, South Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, South Korea
| | - Ki Tae Suk
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, South Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, South Korea
| | - Dong Joon Kim
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, South Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, South Korea
- *Correspondence: Dong Joon Kim,
| |
Collapse
|
129
|
Shi J, Yin Q, Zhang L, Wu Y, Yi P, Guo M, Li H, Yuan L, Wang Z, Zhuang P, Zhang Y. Zi Shen Wan Fang Attenuates Neuroinflammation and Cognitive Function Via Remodeling the Gut Microbiota in Diabetes-Induced Cognitive Impairment Mice. Front Pharmacol 2022; 13:898360. [PMID: 35910371 PMCID: PMC9335489 DOI: 10.3389/fphar.2022.898360] [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: 03/17/2022] [Accepted: 06/02/2022] [Indexed: 11/21/2022] Open
Abstract
Background : Cognitive dysfunction is a critical complication of diabetes mellitus, and there are still no clinically approved drugs. Zi Shen Wan Fang (ZSWF) is an optimized prescription composed of Anemarrhenae Rhizoma, Phellodendri Chinensis Cortex, and Cistanches Herba. The purpose of this study is to investigate the effect of ZSWF on DCI and explore its mechanism from the perspective of maintaining intestinal microbial homeostasis in order to find an effective prescription for treating DCI. Methods: The diabetes model was established by a high-fat diet combined with intraperitoneal injections of streptozotocin (STZ, 120 mg/kg) and the DCI model was screened by Morris water maze (MWM) after 8 weeks of continuous hyperglycemic stimulation. The DCI mice were randomly divided into the model group (DCI), the low- and high-ZSWF-dose groups (9.63 g/kg, 18.72 g/kg), the mixed antibiotic group (ABs), and the ZSWF combined with mixed antibiotic group (ZSWF + ABs). ZSWF was administered orally once a day for 8 weeks. Then, cognitive function was assessed using MWM, neuroinflammation and systemic inflammation were analyzed by enzyme-linked immunosorbent assay kits, intestinal barrier integrity was assessed by hematoxylin-eosin (HE) staining and Western blot and high performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). Furthermore, the alteration to intestinal flora was monitored by 16S rDNA sequencing. Results: ZSWF restored cognitive function in DCI mice and reduced levels of proinflammatory cytokines such as IL-1β, IL-6, and TNF-α. Moreover, ZSWF protected the integrity of the intestinal barrier by increasing intestinal ZO-1 and occludin protein expression and decreasing urinary lactulose to mannitol ratio. In addition, ZSWF reshaped the imbalanced gut microbiota in DCI mice by reversing the abundance changes of a wide range of intestinal bacteria at the phyla and genus levels. In contrast, removing gut microbiota with antibiotics partially eliminated the effects of ZSWF on improving cognitive function and reducing inflammation, confirming the essential role of gut microbiota in the improvement of DCI by ZSWF. Conclusion: ZSWF can reverse cognitive impairment in DCI mice by remolding the structure of destructed gut microbiota community, which is a potential Chinese medicine prescription for DCI treatment.
Collapse
Affiliation(s)
- Jiangwei Shi
- Department of Integrated Rehabilitation, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qingsheng Yin
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Zhang
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yu Wu
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Pengrong Yi
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mengqing Guo
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huhu Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Liuyi Yuan
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zixuan Wang
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Pengwei Zhuang
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanjun Zhang
- Department of Integrated Rehabilitation, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
130
|
Dwaib HS, AlZaim I, Ajouz G, Eid AH, El-Yazbi A. Sex Differences in Cardiovascular Impact of Early Metabolic Impairment: Interplay between Dysbiosis and Adipose Inflammation. Mol Pharmacol 2022; 102:481-500. [PMID: 34732528 DOI: 10.1124/molpharm.121.000338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/23/2021] [Indexed: 11/22/2022] Open
Abstract
The evolving view of gut microbiota has shifted toward describing the colonic flora as a dynamic organ in continuous interaction with systemic physiologic processes. Alterations of the normal gut bacterial profile, known as dysbiosis, has been linked to a wide array of pathologies. Of particular interest is the cardiovascular-metabolic disease continuum originating from positive energy intake and high-fat diets. Accumulating evidence suggests a role for sex hormones in modulating the gut microbiome community. Such a role provides an additional layer of modulation of the early inflammatory changes culminating in negative metabolic and cardiovascular outcomes. In this review, we will shed the light on the role of sex hormones in cardiovascular dysfunction mediated by high-fat diet-induced dysbiosis, together with the possible involvement of insulin resistance and adipose tissue inflammation. Insights into novel therapeutic interventions will be discussed as well. SIGNIFICANCE STATEMENT: Increasing evidence implicates a role for dysbiosis in the cardiovascular complications of metabolic dysfunction. This minireview summarizes the available data on the sex-based differences in gut microbiota alterations associated with dietary patterns leading to metabolic impairment. A role for a differential impact of adipose tissue inflammation across sexes in mediating the cardiovascular detrimental phenotype following diet-induced dysbiosis is proposed. Better understanding of this pathway will help introduce early approaches to mitigate cardiovascular deterioration in metabolic disease.
Collapse
Affiliation(s)
- Haneen S Dwaib
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ghina Ajouz
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ahmed El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| |
Collapse
|
131
|
Zhou L, Li H, Hou G, Wang J, Zhou H, Wang D. Effects of Vine Tea Extract on Meat Quality, Gut Microbiota and Metabolome of Wenchang Broiler. Animals (Basel) 2022; 12:ani12131661. [PMID: 35804560 PMCID: PMC9265100 DOI: 10.3390/ani12131661] [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: 05/16/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
This study investigates the effects of vine tea (Ampelopsis grossedentata) extract (AGE) on meat quality, gut microbiota and cecal content metabolites of Wenchang broilers. A total of 240 female Wenchang broilers aged 70 days were randomly allocated into four groups with five replicates of twelve broilers each. Broilers were fed a corn-soybean basal diet supplemented with AGE at 0 (T1), 0.2% (T2), 0.4% (T3) and 0.6% (T4) until 124 days of age. The whole feeding trial lasted 54 days. Results suggest that the content of total triglycerides and low-density lipoprotein cholesterol in serum of broilers are linearly reduced with dietary AGE supplementation (p < 0.05). The T3 and T4 groups had higher (p < 0.05) a* value in thigh and breast muscles than the T1 group. Additionally, the dietary supplementation of AGE decreased the shear force and drip loss of both thigh and breast muscles linearly (p < 0.05). Compared with the T1 group, AGE supplementation increased the levels of inosine monophosphate (IMP) significantly (p < 0.05) in both the thigh and breast muscles. Furthermore, an increase (p < 0.05) in the total unsaturated fatty acid (USFA), polyunsaturated fatty acids (PUFA) and the ratio of unsaturated fatty acids to saturated fatty acid (USFA: SFA) in both the thigh and breast muscles in the T3 group was observed. Higher abundance of Bacteroidota (p < 0.05) and lower abundance of Firmicutes (p < 0.05) were observed in the T3 group. The abundance of Faecalibacterium was significantly decreased (p < 0.05) in the T3 group compared with the T1 group. Cholesterol sulfate and p-cresol sulfate were identified as differential metabolites between the T1 and T3 groups. It suggested that 0.4% of AGE supplementation significantly downregulated the levels of p-cresol sulfate and cholesterol sulfate (p < 0.05) and the hepatic 3-hydroxy-3-methylglutaryl-CoA reductase (HMGR) activity compared with the control. Our present study demonstrates that dietary supplementation with AGE can improve the quality and flavor by increasing the IMP and PUFA content in the muscle of Wenchang broilers. Furthermore, dietary AGE supplementation with 0.4% can regulate the cholesterol metabolism of Wenchang broilers.
Collapse
Affiliation(s)
- Luli Zhou
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; (L.Z.); (G.H.)
| | - Hui Li
- College of Animal Science and Technology, Hainan University, Haikou 570228, China; (H.L.); (J.W.)
| | - Guanyu Hou
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; (L.Z.); (G.H.)
| | - Jian Wang
- College of Animal Science and Technology, Hainan University, Haikou 570228, China; (H.L.); (J.W.)
| | - Hanlin Zhou
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China
- Correspondence: (H.Z.); (D.W.)
| | - Dingfa Wang
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; (L.Z.); (G.H.)
- Correspondence: (H.Z.); (D.W.)
| |
Collapse
|
132
|
Sung JCC, Lai NCY, Wu KC, Choi MC, Ma CHY, Lin J, Kuok CN, Leong WL, Lam WK, Hamied YK, Lam DMK, Sze ETP, Kwong KWY. Safety and Immunogenicity of Inactivated Bacillus subtilis Spores as a Heterologous Antibody Booster for COVID-19 Vaccines. Vaccines (Basel) 2022; 10:1014. [PMID: 35891178 PMCID: PMC9322707 DOI: 10.3390/vaccines10071014] [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: 05/20/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 12/04/2022] Open
Abstract
The coronavirus diseases 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections have threatened the world for more than 2 years. Multiple vaccine candidates have been developed and approved for emergency use by specific markets, but multiple doses are required to maintain the antibody level. Preliminary safety and immunogenicity data about an oral dose vaccine candidate using recombinant Bacillus subtilis in healthy adults were reported previously from an investigator-initiated trial in Hong Kong. Additional data are required in order to demonstrate the safety and efficacy of the candidate as a heterologous booster in vaccinated recipients. In an ongoing, placebo-controlled, observer-blinded, fixed dose, investigator-initiated trial conducted in the Macau, we randomly assigned healthy adults, 21 to 62 years of age to receive either placebo or a Bacillus subtilis oral dose vaccine candidate, which expressed the spike protein receptor binding domain of SARS-CoV-2 on the spore surface. The primary outcome was safety (e.g., local and systemic reactions and adverse events); immunogenicity was a secondary outcome. For both the active vaccine and placebo, participants received three courses in three consecutive days. A total of 16 participants underwent randomization: 9 participants received vaccine and 7 received placebo. No observable local or systemic side-effect was reported. In both younger and older adults receiving placebo, the neutralizing antibody levels were gradually declining, whereas the participants receiving the antibody booster showed an increase in neutralizing antibody level.
Collapse
Affiliation(s)
- Johnny Chun-Chau Sung
- Research Department, DreamTec Cytokines Limited, Hong Kong, China; (J.C.-C.S.); (N.C.-Y.L.); (K.-C.W.); (M.-C.C.); (C.H.-Y.M.); (J.L.)
- Oristry BioTech (HK) Limited, Hong Kong, China
- Meserna Therapeutic (HK) Limited, Hong Kong, China
- L&L Immunotherapy Company Limited, Hong Kong, China;
| | - Nelson Cheuk-Yin Lai
- Research Department, DreamTec Cytokines Limited, Hong Kong, China; (J.C.-C.S.); (N.C.-Y.L.); (K.-C.W.); (M.-C.C.); (C.H.-Y.M.); (J.L.)
- Oristry BioTech (HK) Limited, Hong Kong, China
- Meserna Therapeutic (HK) Limited, Hong Kong, China
- L&L Immunotherapy Company Limited, Hong Kong, China;
| | - Kam-Chau Wu
- Research Department, DreamTec Cytokines Limited, Hong Kong, China; (J.C.-C.S.); (N.C.-Y.L.); (K.-C.W.); (M.-C.C.); (C.H.-Y.M.); (J.L.)
| | - Man-Chung Choi
- Research Department, DreamTec Cytokines Limited, Hong Kong, China; (J.C.-C.S.); (N.C.-Y.L.); (K.-C.W.); (M.-C.C.); (C.H.-Y.M.); (J.L.)
| | - Chloe Ho-Yi Ma
- Research Department, DreamTec Cytokines Limited, Hong Kong, China; (J.C.-C.S.); (N.C.-Y.L.); (K.-C.W.); (M.-C.C.); (C.H.-Y.M.); (J.L.)
| | - Jayman Lin
- Research Department, DreamTec Cytokines Limited, Hong Kong, China; (J.C.-C.S.); (N.C.-Y.L.); (K.-C.W.); (M.-C.C.); (C.H.-Y.M.); (J.L.)
- L&L Immunotherapy Company Limited, Hong Kong, China;
| | - Cheong-Nang Kuok
- Macao Greater Bay Area Association of Healthcare Providers, Macau 999078, China; (C.-N.K.); (W.-L.L.); (W.-K.L.)
| | - Wai-Leng Leong
- Macao Greater Bay Area Association of Healthcare Providers, Macau 999078, China; (C.-N.K.); (W.-L.L.); (W.-K.L.)
| | - Weng-Kei Lam
- Macao Greater Bay Area Association of Healthcare Providers, Macau 999078, China; (C.-N.K.); (W.-L.L.); (W.-K.L.)
| | | | - Dominic Man-Kit Lam
- L&L Immunotherapy Company Limited, Hong Kong, China;
- Torsten Wiesel International Research Institute, Sichuan University, Chengdu 610064, China
| | - Eric Tung-Po Sze
- School of Science and Technology, Hong Kong Metropolitan University, Hong Kong, China;
| | - Keith Wai-Yeung Kwong
- Research Department, DreamTec Cytokines Limited, Hong Kong, China; (J.C.-C.S.); (N.C.-Y.L.); (K.-C.W.); (M.-C.C.); (C.H.-Y.M.); (J.L.)
- Oristry BioTech (HK) Limited, Hong Kong, China
- Meserna Therapeutic (HK) Limited, Hong Kong, China
- L&L Immunotherapy Company Limited, Hong Kong, China;
| |
Collapse
|
133
|
Effects of Spore-Displayed p75 Protein from Lacticaseibacillus rhamnosus GG on the Transcriptional Response of HT-29 Cells. Microorganisms 2022; 10:microorganisms10071276. [PMID: 35888995 PMCID: PMC9323162 DOI: 10.3390/microorganisms10071276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/10/2022] Open
Abstract
A Lacticaseibacillus rhamnosus GG-derived protein, p75, is one of the key molecules exhibiting probiotic activity. However, the molecular mechanism and transcriptional response of p75 in human intestinal epithelial cells are not completely understood. To gain a deeper understanding of its potential probiotic action, this study investigated genome-wide responses of HT-29 cells to stimulation by spore-displayed p75 (CotG-p75) through a transcriptome analysis based on RNA sequencing. Analysis of RNA-seq data showed significant changes of gene expression in HT-29 cells stimulated by CotG-p75 compared to the control. A total of 189 up-regulated and 314 down-regulated genes was found as differentially expressed genes. Gene ontology enrichment analysis revealed that a large number of activated genes was involved in biological processes, such as epithelial cell differentiation, development, and regulation of cell proliferation. A gene–gene interaction network analysis showed that several DEGs, including AREG, EREG, HBEGF, EPGN, FASLG, GLI2, CDKN1A, FOSL1, MYC, SERPINE1, TNFSF10, BCL6, FLG, IVL, SPRR1A, SPRR1B, SPRR3, and MUC5AC, might play a critical role in these biological processes. RNA-seq results for selected genes were verified by reverse transcription-quantitative polymerase chain reaction. Overall, these results provide extensive knowledge about the transcriptional responses of HT-29 cells to stimulation by CotG-p75. This study showed that CotG-p75 can contribute to cell survival and epithelial development in human intestinal epithelial cells.
Collapse
|
134
|
Rahman MM, Islam F, -Or-Rashid MH, Mamun AA, Rahaman MS, Islam MM, Meem AFK, Sutradhar PR, Mitra S, Mimi AA, Emran TB, Fatimawali, Idroes R, Tallei TE, Ahmed M, Cavalu S. The Gut Microbiota (Microbiome) in Cardiovascular Disease and Its Therapeutic Regulation. Front Cell Infect Microbiol 2022; 12:903570. [PMID: 35795187 PMCID: PMC9251340 DOI: 10.3389/fcimb.2022.903570] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/09/2022] [Indexed: 12/11/2022] Open
Abstract
In the last two decades, considerable interest has been shown in understanding the development of the gut microbiota and its internal and external effects on the intestine, as well as the risk factors for cardiovascular diseases (CVDs) such as metabolic syndrome. The intestinal microbiota plays a pivotal role in human health and disease. Recent studies revealed that the gut microbiota can affect the host body. CVDs are a leading cause of morbidity and mortality, and patients favor death over chronic kidney disease. For the function of gut microbiota in the host, molecules have to penetrate the intestinal epithelium or the surface cells of the host. Gut microbiota can utilize trimethylamine, N-oxide, short-chain fatty acids, and primary and secondary bile acid pathways. By affecting these living cells, the gut microbiota can cause heart failure, atherosclerosis, hypertension, myocardial fibrosis, myocardial infarction, and coronary artery disease. Previous studies of the gut microbiota and its relation to stroke pathogenesis and its consequences can provide new therapeutic prospects. This review highlights the interplay between the microbiota and its metabolites and addresses related interventions for the treatment of CVDs.
Collapse
|
135
|
Wang K, Yang A, Peng X, Lv F, Wang Y, Cui Y, Wang Y, Zhou J, Si H. Linkages of Various Calcium Sources on Immune Performance, Diarrhea Rate, Intestinal Barrier, and Post-gut Microbial Structure and Function in Piglets. Front Nutr 2022; 9:921773. [PMID: 35782941 PMCID: PMC9248811 DOI: 10.3389/fnut.2022.921773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/20/2022] [Indexed: 01/08/2023] Open
Abstract
The purpose of this experiment was to investigate the effects of different sources of calcium on immune performance, diarrhea rate, intestinal barrier, and post-intestinal flora structure and function in weaned piglets. A total of 1,000 weaned piglets were randomly assigned to five groups 10 replicate pens per treatment, 20 piglets per pen and fed calcium carbonate, calcium citrate, multiple calcium, and organic trace minerals of different concentrations of acidifier diets. The results of the study showed that the replacement of calcium carbonate with calcium citrate and multiple calcium had almost no significant effect on immune indexes (IL-1β, IL-6, IL-10, TNF-α) of piglets compared with the control group (p > 0.05). The five groups did not show a change in the diarrhea rate and diarrhea index (p > 0.05). The diet containing multiple calcium dramatically decreased the TP compared to the C and L diet (p < 0.05). No significant difference in HDL was noted in the five groups (p > 0.05). However, the concentration of LDL in blood in the multiple calcium group was significantly higher than that in groups L and D (p < 0.05). Moreover, the concentration of Glu in blood in the multiple calcium group was significantly higher than that in group C (p < 0.05). Compared with the control group, calcium citrate plus organic trace minerals diet markedly increased UCG-005 abundance in the colon (p < 0.05). In addition, the relative abundance of Prevotellaceae_NK3B31_group had an upward trend in the colon of the M group compared to the D group (p = 0.070). Meanwhile, calcium citrate plus organic trace minerals diet markedly increased Clostridium_sensu_stricto_1 abundance in the colon (p < 0.05). Metagenomic predictions by PICRUSt suggested that the colonic and fecal microbiota was mainly involved in carbohydrate metabolism, amino acid metabolism, energy metabolism, and metabolism of cofactors and vitamins.
Collapse
Affiliation(s)
- Kaijun Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Anqi Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Xiaomin Peng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Feifei Lv
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Ying Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yao Cui
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yuhan Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | | | - Hongbin Si
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- *Correspondence: Hongbin Si,
| |
Collapse
|
136
|
Wang K, Peng X, Yang A, Huang Y, Tan Y, Qian Y, Lv F, Si H. Effects of Diets With Different Protein Levels on Lipid Metabolism and Gut Microbes in the Host of Different Genders. Front Nutr 2022; 9:940217. [PMID: 35782952 PMCID: PMC9240812 DOI: 10.3389/fnut.2022.940217] [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: 05/10/2022] [Accepted: 05/30/2022] [Indexed: 12/21/2022] Open
Abstract
The purpose of this experiment was to investigate the effects of different protein levels on lipid metabolism and gut microbes in mice of different genders. A total of 60 mice (30 female and 30 male) were randomly assigned to six groups and fed female mice with low protein diet (FLP), basal protein diet (FBD), and high protein diet (FHP). Similarly, the male mice fed with low protein diet (MLP), basal protein diet (MBD), and high protein diet (MHP). The low protein diet contained 14% CP, the basal diet contained 20% CP, and the high protein diet contained 26% CP. The results of the study showed that both basal and high protein diets significantly reduced the perirenal adipose tissues (PEAT) index in male mice compared to low protein diet (p < 0.05). For the gut, the FHP significantly increased the relative gut weight compared to the FBD and FLP (p < 0.05). At the same time, the FHP also significantly increased the relative gut length compared with the FBD and FLP (p < 0.05). The MHP significantly increased TC concentration compared with the MLP (p < 0.05), and the MBD tended to increase TC concentration compared with the MLP in serum (p = 0.084). The histomorphology result of the jejunum and ileum showed that a low protein diet was beneficial to the digestion and absorption of nutrients in the small intestine of mice. While different protein levels had no effect on the total number of fecal microbial species in mice, different protein levels had a significant effect on certain fecal microbes in mice, the absolute abundance of Verrucomicrobia in the feces of male mice was significantly higher in both high and basal protein diets than in the low protein diet (p < 0.05). The high protein diet significantly reduced the absolute abundance of Patescibacteria in the feces of female mice compared to both the basal and low protein diets (p < 0.05). The absolute abundance of Patescibacteria in male feces was not affected by dietary protein levels (p > 0.05). Taken together, our results suggest that a low protein diet can alter fat deposition and lipid metabolism in mice, and that it benefited small intestinal epithelial structure and microbes.
Collapse
Affiliation(s)
- Kaijun Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xiaomin Peng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Anqi Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yiqin Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yuxiao Tan
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yajing Qian
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Feifei Lv
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Hongbin Si
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- *Correspondence: Hongbin Si,
| |
Collapse
|
137
|
Li Y, Bi Y, Yang L, Jin K. Comparative study on intestinal microbiome composition and function in young and adult Hainan gibbons ( Nomascus hainanus). PeerJ 2022; 10:e13527. [PMID: 35698614 PMCID: PMC9188309 DOI: 10.7717/peerj.13527] [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: 03/15/2022] [Accepted: 05/11/2022] [Indexed: 01/17/2023] Open
Abstract
The Hainan gibbon is one of the most endangered primates in the world, with a small population size, narrow distribution range, and high inbreeding risk, which retains the risk of species extinction. To explore the composition and functional differences of the intestinal microbiome of Hainan gibbons at different ages, the faecal microbiomes of young and adult Hainan gibbons were analysed using metagenome sequencing. The results showed that the dominant phyla in the intestinal tract of young and adult Hainan gibbons were Firmicutes and Bacteroidetes, and the dominant genus was Prevotella. Linear discriminant analysis effect size analysis showed that Firmicutes, Ruminococcus, Clostridium, and Butyrivibrio were significantly more abundant in adults than in young, whereas Bacteroidetes, Proteobacteria, Prevotella, and Bacteroides were significantly more abundant in young than in adults. In terms of gene function, the adult Hainan gibbon intestinal microbiome generally harboured a higher abundance of genes related to metabolic processes, such as carbohydrate, amino acid, and nucleotide metabolism. This may be due to adaptive advantages for adult Hainan gibbons, such as stable and mature intestinal microbiome composition, which allows them to utilise diverse foods efficiently. In summary, this study helps understand the dynamic changes in the intestinal microbiome of young and adult Hainan gibbons and plays a key role in the health monitoring and rejuvenation of their population.
Collapse
Affiliation(s)
- Yimeng Li
- Institute of Forest Ecology-Environment and Nature Conservation, Chinese Academy of Forestry, Beijing, China,Beijing Museum of Natural History, Beijing, China,Hainan Institute of National Park, Haikou, China
| | - Yu Bi
- Institute of Forest Ecology-Environment and Nature Conservation, Chinese Academy of Forestry, Beijing, China,Hainan Institute of National Park, Haikou, China,Research Institute of Natural Protected Area, Chinese Academy of Forestry, Beijing, China,Key Laboratory of Biodiversity Conservation of National Forestry and Grassland Administration, Beijing, China
| | - Liangliang Yang
- Institute of Forest Ecology-Environment and Nature Conservation, Chinese Academy of Forestry, Beijing, China,Key Laboratory of Biodiversity Conservation of National Forestry and Grassland Administration, Beijing, China
| | - Kun Jin
- Institute of Forest Ecology-Environment and Nature Conservation, Chinese Academy of Forestry, Beijing, China,Hainan Institute of National Park, Haikou, China,Research Institute of Natural Protected Area, Chinese Academy of Forestry, Beijing, China,Key Laboratory of Biodiversity Conservation of National Forestry and Grassland Administration, Beijing, China
| |
Collapse
|
138
|
Effects of Vitamin A on Yanbian Yellow Cattle and Their Preadipocytes by Activating AKT/mTOR Signaling Pathway and Intestinal Microflora. Animals (Basel) 2022; 12:ani12121477. [PMID: 35739812 PMCID: PMC9219514 DOI: 10.3390/ani12121477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Vitamin A is a fat-soluble vitamin that not only plays a role in vision, growth, and development, but also in fat production and metabolism in animals. To improve the production of high-grade beef, it is necessary to explore the molecular mechanism of intramuscular fat deposition in beef cattle through molecular biology techniques. In this study, we selected Yanbian yellow cattle, one of the five major cattle breeds in China, to investigate the effects of vitamin A and its metabolite, all-trans retinoic acid (ATRA), on the proliferation and differentiation of preadipocytes and changes in intestinal microorganisms. It was found that ATRA inhibited adipogenic differentiation of preadipocytes in Yanbian yellow cattle via the AKT/mTOR signaling pathway. This study provides insight into nutritional management and reveals the role of vitamin A in lipid metabolism in Yanbian yellow cattle. Abstract In this study, the effects of vitamin A and its metabolite, all-trans retinoic acid (ATRA), on the proliferation and differentiation of preadipocytes and the intestinal microbiome in Yanbian yellow cattle were investigated. Preadipocytes collected from Yanbian yellow cattle treated with different concentrations of ATRA remained in the G1/G0 phase, as determined by flow cytometry. Quantitative reverse-transcription polymerase chain reaction and western blotting analyses showed that the mRNA and protein expression levels of key adipogenic factors, peroxisome proliferator- activated receptor gamma (PPARγ), CCAAT enhancer-binding protein α (C/EBPα), and extracellular signal-regulated kinase 2 (ERK2), decreased. ATRA was found to regulate the mTOR signaling pathway, which is involved in lipid metabolism, by inhibiting the expression of AKT2 and the adipogenic transcription factors SREBP1, ACC, and FAS; the protein and mRNA expression levels showed consistent trends. In addition, 16S rRNA sequencing results showed that a low concentration of vitamin A promoted the growth of intestinal microflora beneficial to lipid metabolism and maintained intestinal health. The results indicated that ATRA inhibited the adipogenic differentiation of preadipocytes from Yanbian yellow cattle through the AKT/mTOR signaling pathway, and that low concentrations of vitamin A may help maintain the intestinal microbes involved in lipid metabolism in cattle.
Collapse
|
139
|
Baholet D, Skalickova S, Batik A, Malyugina S, Skladanka J, Horky P. Importance of Zinc Nanoparticles for the Intestinal Microbiome of Weaned Piglets. Front Vet Sci 2022; 9:852085. [PMID: 35720843 PMCID: PMC9201420 DOI: 10.3389/fvets.2022.852085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
The scientific community is closely monitoring the replacement of antibiotics with doses of ZnO in weaned piglets. Since 2022, the use of zinc in medical doses has been banned in the European Union. Therefore, pig farmers are looking for other solutions. Some studies have suggested that zinc nanoparticles might replace ZnO for the prevention of diarrhea in weaning piglets. Like ZnO, zinc nanoparticles are effective against pathogenic microorganisms, e.g., Enterobacteriaceae family in vitro and in vivo. However, the effect on probiotic Lactobacillaceae appears to differ for ZnO and zinc nanoparticles. While ZnO increases their numbers, zinc nanoparticles act in the opposite way. These phenomena have been also confirmed by in vitro studies that reported a strong antimicrobial effect of zinc nanoparticles against Lactobacillales order. Contradictory evidence makes this topic still controversial, however. In addition, zinc nanoparticles vary in their morphology and properties based on the method of their synthesis. This makes it difficult to understand the effect of zinc nanoparticles on the intestinal microbiome. This review is aimed at clarifying many circumstances that may affect the action of nanoparticles on the weaning piglets' microbiome, including a comprehensive overview of the zinc nanoparticles in vitro effects on bacterial species occurring in the digestive tract of weaned piglets.
Collapse
Affiliation(s)
- Daria Baholet
- Department of Animal Nutrition and Forage Production, Mendel University in Brno, Brno, Czechia
| | - Sylvie Skalickova
- Department of Animal Nutrition and Forage Production, Mendel University in Brno, Brno, Czechia
| | - Andrej Batik
- Department of Animal Morphology, Physiology and Genetics, Mendel University in Brno, Brno, Czechia
| | - Svetlana Malyugina
- Department of Animal Nutrition and Forage Production, Mendel University in Brno, Brno, Czechia
| | - Jiri Skladanka
- Department of Animal Nutrition and Forage Production, Mendel University in Brno, Brno, Czechia
| | - Pavel Horky
- Department of Animal Nutrition and Forage Production, Mendel University in Brno, Brno, Czechia
- *Correspondence: Pavel Horky
| |
Collapse
|
140
|
Layunta E, Forcén R, Grasa L. TLR2 and TLR4 Modulate Mouse Ileal Motility by the Interaction with Muscarinic and Nicotinic Receptors. Cells 2022; 11:cells11111791. [PMID: 35681486 PMCID: PMC9180263 DOI: 10.3390/cells11111791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a chronic functional bowel disorder characterized by intestinal dysmotility. Changes in intestinal microbiota (dysbiosis) can lead to alterations in neuro-muscular functions in the gut. Toll-like receptors (TLRs) 2 and 4 recognize intestinal bacteria and are involved in the motor response induced by gastrointestinal (GI) neurotransmitters. Acetylcholine (ACh) is a well-known neurotransmitter involved in the regulation of GI motility. This study aimed to evaluate the role of TLR2 and TLR4 in the intestinal motor-response induced by ACh in the mouse ileum, as well as the expression and function of the muscarinic and nicotinic ACh receptors. Muscle contractility studies showed that the contractions induced by ACh were significantly lower in TLR2−/− and TLR4−/− with respect to WT mice. In WT mice, the contractions induced by ACh were reduced in the presence of AF-DX AF-DX 116 (a muscarinic ACh receptor (mAChR) M2 antagonist), 4-DAMP (a mAChR M3 antagonist), mecamylamine (a nicotinic AChR receptor (nAChR) α3β4 antagonist) and α-bungarotoxin (a nAChR α7 antagonist). In TLR2−/− mice, the contractions induced by ACh were increased by AF-DX 116 and mecamylamine. In TLR4−/− mice, the contractions induced by ACh were reduced by α-bungarotoxin and 4-DAMP. The mRNA and protein expressions of M3 and α3 receptors were diminished in the ileum from TLR2−/− and TLR4−/− with respect to WT mice. However, the levels of mRNA and protein of β4 were diminished only in TLR4−/− but not in TLR2−/− mice. In conclusion, our results show that TLR2 and TLR4 modulates the motor responses to ACh in the mouse ileum. TLR2 acts on muscarinic M2 and M3 and nicotinic α3β4 ACh receptors, while TLR4 acts on muscarinic M3 and nicotinic α3β4 and α7 ACh receptors.
Collapse
Affiliation(s)
- Elena Layunta
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Medicinaregatan 9C, 41390 Gothenburg, Sweden;
| | - Raquel Forcén
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain;
| | - Laura Grasa
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain;
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Instituto Agroalimentario de Aragón—IA2—(Universidad de Zaragoza-CITA), 50013 Zaragoza, Spain
- Correspondence:
| |
Collapse
|
141
|
Tian Z, Pu H, Cai D, Luo G, Zhao L, Li K, Zou J, Zhao X, Yu M, Wu Y, Yang T, Guo P, Hu X. Characterization of the bacterial microbiota in different gut and oral compartments of splendid japalure (Japalura sensu lato). BMC Vet Res 2022; 18:205. [PMID: 35624481 PMCID: PMC9137078 DOI: 10.1186/s12917-022-03300-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/16/2022] [Indexed: 11/10/2022] Open
Abstract
Background Gut and oral microbes form complex communities and play key roles in co-evolution with their hosts. However, little is understood about the bacterial community in lizards. Results In this study, we investigated the gut and oral bacterial communities in Japalura sensu lato from Sichuan Province, China, using 16S rRNA gene sequencing. Results showed that Bacteroidota (36.5%) and Firmicutes (32.8%) were the main phyla in the gut, while Proteobacteria, Bacteroidota, Firmicutes, and Actinobacteriota were the dominant phyla in the oral cavity. 16 S rRNA sequencing analysis of fecal samples showed that: (1) Bacteroidota was the most abundant in Japalura sensu lato, which was different from the bacterial community of insectivorous animals; (2) Bacteroidota, Firmicutes, Actinobacteriota, Fusobacteriota, and Cyanobacteria were the most abundant phylum in Japalura sensu lato. (3) Proteobacteria was the dominant phylum in Japalura sensu lato and other domestic insectivorous lizards (Shinisaurus crocodilurus, Phrynocephalus vlangalii, and Takydromus septentrionalis); (4) Comparing with the bacterial community of Shinisaurus crocodilurus, Phrynocephalus vlangalii, Takydromus septentrionalis, Liolaemus parvus, L. ruibali, and Phymaturus williamsi, Desulfobacterota was uniquely present in the gut of Japalura sensu lato. 16 S rRNA sequencing of oral samples showed that Chloroflexi and Deinococcota phyla were enriched in the oral cavity, which may have a significant influence on living in extreme environments. Conclusions Thus, based on 16 S rRNA sequencing analysis of the community composition of the gut and oral microbiomes, this study firstly represents a foundation for understanding the gut and oral microbial ecology of Japalura sensu lato, and constitutes a detail account of the diversity of the microbiota inhabiting the gut and oral cavity of Japalura sensu lato. Further researches will continue to reveal how gut and oral microbial communities may be impacting the ecology and evolution of lizards. Supplementary information The online version contains supplementary material available at 10.1186/s12917-022-03300-w.
Collapse
Affiliation(s)
- Zhige Tian
- Faculty of Agriculture, Forestry and Food Engineering, Yibin Key Laboratory of Zoological Diversity and Ecological Conservation, Yibin University, 644000, Yibin, People's Republic of China
| | - Hongli Pu
- Faculty of Agriculture, Forestry and Food Engineering, Yibin Key Laboratory of Zoological Diversity and Ecological Conservation, Yibin University, 644000, Yibin, People's Republic of China
| | - Dongdong Cai
- Sichuan Animal Disease Control Central, 610000, Chengdu, People's Republic of China
| | - Guangmei Luo
- Faculty of Agriculture, Forestry and Food Engineering, Yibin Key Laboratory of Zoological Diversity and Ecological Conservation, Yibin University, 644000, Yibin, People's Republic of China
| | - Lili Zhao
- College of Veterinary Medicine, Jilin University, 130000, Changchun, People's Republic of China
| | - Ke Li
- Faculty of Agriculture, Forestry and Food Engineering, Yibin Key Laboratory of Zoological Diversity and Ecological Conservation, Yibin University, 644000, Yibin, People's Republic of China
| | - Jie Zou
- Faculty of Agriculture, Forestry and Food Engineering, Yibin Key Laboratory of Zoological Diversity and Ecological Conservation, Yibin University, 644000, Yibin, People's Republic of China
| | - Xiang Zhao
- Faculty of Agriculture, Forestry and Food Engineering, Yibin Key Laboratory of Zoological Diversity and Ecological Conservation, Yibin University, 644000, Yibin, People's Republic of China
| | - Min Yu
- Faculty of Agriculture, Forestry and Food Engineering, Yibin Key Laboratory of Zoological Diversity and Ecological Conservation, Yibin University, 644000, Yibin, People's Republic of China
| | - Yayong Wu
- Faculty of Agriculture, Forestry and Food Engineering, Yibin Key Laboratory of Zoological Diversity and Ecological Conservation, Yibin University, 644000, Yibin, People's Republic of China
| | - Tiankuo Yang
- Aviation Medical Appraisal Center, Civil Aviation Flight University of China, 618307, Guanghan, China.
| | - Peng Guo
- Faculty of Agriculture, Forestry and Food Engineering, Yibin Key Laboratory of Zoological Diversity and Ecological Conservation, Yibin University, 644000, Yibin, People's Republic of China.
| | - Xiaoliang Hu
- Faculty of Agriculture, Forestry and Food Engineering, Yibin Key Laboratory of Zoological Diversity and Ecological Conservation, Yibin University, 644000, Yibin, People's Republic of China.
| |
Collapse
|
142
|
Interaction Between Altered Gut Microbiota and Sepsis: A Hypothesis or an Authentic Fact? J Intensive Care Med 2022; 38:121-131. [DOI: 10.1177/08850666221102796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sepsis, as an important public health concern, is one of the leading causes of death in hospitals around the world, accounting for 25% of all deaths. Nowadays, several factors contribute to the development of sepsis. The role of the gut microbiota and the response state of the aberrant immune system is dominant. The effect of the human microbiome on health is undeniable, and gut microbiota is even considered a body organ. It is now clear that the alteration in the normal balance of the microbiota (dysbiosis) is associated with a change in the status of immune system responses. Owing to the strong association between the gut microbiota and its metabolites particularly short-chain fatty acids with many illnesses, the gut microbiota has a unique position in the research of microbiologists and even clinicians. This review aimed to analyze studies’ results on the association between microbiota and sepsis, with a substantial understanding of their relationship. As a result, an extensive and comprehensive search was conducted on this issue in existing databases.
Collapse
|
143
|
Yang A, Wang K, Peng X, Lv F, Wang Y, Cui Y, Wang Y, Qu D, Zhou J, Si H. Effects of Different Sources of Calcium in the Diet on Growth Performance, Blood Metabolic Parameters, and Intestinal Bacterial Community and Function of Weaned Piglets. Front Nutr 2022; 9:885497. [PMID: 35571955 PMCID: PMC9101144 DOI: 10.3389/fnut.2022.885497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/31/2022] [Indexed: 12/27/2022] Open
Abstract
Despite a well-documented effect of calcium on the piglet's intestinal microbiota composition, it is less known about changes in microbial function or the effect of different sources of calcium. The experiment was designed to study the effects of dietary calcium from different sources on production, immune indexes, antioxidant capacity, serum biochemical indexes, and intestinal microflora of weaning piglets. A total of 1,000 piglets were randomly assigned to five groups (10 replicate pens per treatment with 20 pigs per pen) and fed diets supplemented with calcium carbonate, calcium citrate, multiple calcium, organic trace minerals, and different concentrations of acidifier. The results showed that the replacement of calcium carbonate with calcium citrate and multiple calcium had almost no significant difference in the growth performance of pigs compared with the control group, and only the diet of multiple calcium dramatically decreased the average daily feed intake (ADFI) compared to the calcium citrate diet on days 15-28 (p < 0.05). The five groups did not change the content of MDA, SOD, and GSH-Px (p > 0.10). A similar situation occurs in the immune function of the blood. There was no significant effect in immune indexes (IgA, IgG, and IgM) among different treatments after weaning at 6 weeks for piglets (p > 0.10). The 16S rRNA sequencing of ileal and cecal microbiota revealed that only the relative abundance of Actinobacteriota at the phyla level was significantly greater in the ileum of the A group compared to the other treatments (p < 0.05). There was a clear effect on seven bacteria in the top 30 genera of ileum and cecum for five groups (p < 0.05). The result of PICRUSt predicted that the intestinal microbe was mainly involved in carbohydrate and amino acid metabolism, membrane transport, and metabolism of cofactors and vitamins. Besides, adding calcium citrate to a weaned piglet diet is better than other choices from the third week to the fourth week. In conclusion, diets with different calcium sources changed ADFI and some intestinal microbial composition of weaned piglets but had little effect on intestinal microbial function.
Collapse
Affiliation(s)
- Anqi Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Kaijun Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China.,Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xiaomin Peng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Feifei Lv
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Ying Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yao Cui
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yuhan Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Dongshuai Qu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | | | - Hongbin Si
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| |
Collapse
|
144
|
Yan J, Chen Q, Tian L, Li K, Lai W, Bian L, Han J, Jia R, Liu X, Xi Z. Intestinal toxicity of micro- and nano-particles of foodborne titanium dioxide in juvenile mice: Disorders of gut microbiota-host co-metabolites and intestinal barrier damage. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 821:153279. [PMID: 35074372 DOI: 10.1016/j.scitotenv.2022.153279] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/09/2022] [Accepted: 01/16/2022] [Indexed: 05/28/2023]
Abstract
The wide use of TiO2 particles in food and the high exposure risk to children have prompted research into the health risks of TiO2. We used the microbiome and targeted metabolomics to explore the potential mechanism of intestinal toxicity of foodborne TiO2 micro-/nanoparticles after oral exposure for 28 days in juvenile mice. Results showed that the gut microbiota-including the abundance of Bacteroides, Bifidobacterium, Lactobacillus, and Prevotella-changed dynamically during exposure. The organic inflammatory response was activated, and lipopolysaccharide levels increased. Intestinal toxicity manifested as increased mucosal permeability, impaired intestinal barrier, immune damage, and pathological changes. The expression of antimicrobial peptides, occludin, and ZO-1 significantly reduced, while that of JNK2 and Src/pSrc increased. Compared with micro-TiO2 particles, the nano-TiO2 particles had strong toxicity. Fecal microbiota transplant confirmed the key role of gut microbiota in intestinal toxicity. The levels of gut microbiota-host co-metabolites, including pyroglutamic acid, L-glutamic acid, phenylacetic acid, and 3-hydroxyphenylacetic acid, changed significantly. Significant changes were observed in the glutathione and propanoate metabolic pathways. There was a significant correlation between the changes in gut microbiota, metabolites, and intestinal cytokine levels. These, together with the intestinal barrier damage signaling pathway, constitute the network mechanism of the intestinal toxicity of TiO2 particles.
Collapse
Affiliation(s)
- Jun Yan
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Qi Chen
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Lei Tian
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Kang Li
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Wenqing Lai
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Liping Bian
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Jie Han
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Rui Jia
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Xiaohua Liu
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China.
| | - Zhuge Xi
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China.
| |
Collapse
|
145
|
Implications of microbe-mediated crosstalk in the gut: Impact on metabolic diseases. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159180. [PMID: 35568374 DOI: 10.1016/j.bbalip.2022.159180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 01/06/2023]
Abstract
Metabolic diseases continue to afflict most of the U.S. population. Advancements in gut microbiota research have led to the discovery of various functional roles of microorganisms that influence the development of obesity and co-morbidities including type 2 diabetes, non-alcoholic fatty liver disease and cardiovascular disease. Many mechanisms behind these host-microbe interactions stem from processes involving the intestinal epithelium including lipid metabolism. Thus, the purpose of this review is to discuss gut microbe-mediated changes in intestinal physiology and lipid metabolism that contribute to obesity, type 2 diabetes, non-alcoholic fatty liver disease and cardiovascular disease. Within each disease state, the causal role of bacteria in both driving disease development and protecting against metabolic disease will be discussed.
Collapse
|
146
|
Yang H, Jin X, Cheng T, Shan G, Lu C, Gu J, Zhan C, Xu F, Ge D. Comprehensive Genome-Scale Analysis of Esophageal Carcinoma With Esophageal Tissue-Resident Micro-Environment Discrepancy. Front Microbiol 2022; 13:859352. [PMID: 35586863 PMCID: PMC9108775 DOI: 10.3389/fmicb.2022.859352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/05/2022] [Indexed: 12/24/2022] Open
Abstract
To figure out the molecular mechanism in the esophageal squamous carcinoma (ESCC) with the discrepancy in the tissue-resident microbiota, we selected clinical features, RNA sequences, and transcriptomes of ESCC patients from The Cancer Genome Atlas (TCGA) website and detailed tissue-resident microbiota information from The Cancer Microbiome Atlas (n = 60) and explored the infiltration condition of particular microbiota in each sample. We classified the tissue-resident micro-environment of ESCC into two clusters (A and B) and built a predictive classifier model. Cluster A has a higher proportion of certain tissue-resident microbiota with comparatively better survival, while Cluster B has a lower proportion of certain tissue-resident microbiota with comparatively worse survival. We showed traits of gene and clinicopathology in the esophageal tissue-resident micro-environment (ETM) phenotypes. By comparing the two clusters’ molecular signatures, we find that the two clusters have obvious differences in gene expression and mutation, which lead to pathway expression discrepancy. Several pathways are closely related to tumorigenesis. Our results may demonstrate a synthesis of the infiltration pattern of the esophageal tissue-resident micro-environment in ESCC. We reveal the mechanism of esophageal tissue-resident microbiota discrepancy in ESCC, which may contribute to therapy progress for patients with ESCC.
Collapse
|
147
|
Clinical and Preclinical Studies of Fermented Foods and Their Effects on Alzheimer’s Disease. Antioxidants (Basel) 2022; 11:antiox11050883. [PMID: 35624749 PMCID: PMC9137914 DOI: 10.3390/antiox11050883] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
The focus on managing Alzheimer’s disease (AD) is shifting towards prevention through lifestyle modification instead of treatments since the currently available treatment options are only capable of providing symptomatic relief marginally and result in various side effects. Numerous studies have reported that the intake of fermented foods resulted in the successful management of AD. Food fermentation is a biochemical process where the microorganisms metabolize the constituents of raw food materials, giving vastly different organoleptic properties and additional nutritional value, and improved biosafety effects in the final products. The consumption of fermented foods is associated with a wide array of nutraceutical benefits, including anti-oxidative, anti-inflammatory, neuroprotective, anti-apoptotic, anti-cancer, anti-fungal, anti-bacterial, immunomodulatory, and hypocholesterolemic properties. Due to their promising health benefits, fermented food products have a great prospect for commercialization in the food industry. This paper reviews the memory and cognitive enhancement and neuroprotective potential of fermented food products on AD, the recently commercialized fermented food products in the health and food industries, and their limitations. The literature reviewed here demonstrates a growing demand for fermented food products as alternative therapeutic options for the prevention and management of AD.
Collapse
|
148
|
Han LX, Yao WL, Pan J, Wang BS, He WH, Fan XP, Wang WH, Zhang WD. Moniezia benedeni Infection Restrain IgA+, IgG+, and IgM+ Cells Residence in Sheep (Ovis aries) Small Intestine. Front Vet Sci 2022; 9:878467. [PMID: 35573414 PMCID: PMC9096708 DOI: 10.3389/fvets.2022.878467] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/07/2022] [Indexed: 01/12/2023] Open
Abstract
Secreted immunoglobulin A (SIgA), IgG, and IgM play a crucial role in forming the intestinal mucosal immune barrier, and parasites could disturb the host's immune response by releasing various immunomodulatory molecules. Moniezia benedeni is an important pathogen parasitizing in the sheep small intestine. It is aimed to explore the residence characteristics of IgA+, IgG+, and IgM+ cells in the sheep small intestine, and the influence of Moniezia benedeni infection on them. Control group (n = 6) and infected group (n = 6) were selected, respectively, and the three subtype cells residing in the small intestine were systematically observed and analyzed. The results showed that in the Control group, the three types of positive cells were all distributed diffusely, and the total densities in jejunum, duodenum and ileum was gradually declined in turn. Notably, the change trend of IgA+ and IgG+ cells densities were both congruent with the total densities, and the differences among them were significant, respectively (P < 0.05); the IgM+ cells density was the highest in duodenum, followed by jejunum and ileum, there was no significant difference between duodenum and jejunum (P > 0.05), but both significantly higher than in ileum (P < 0.05). In the Infected group, their total densities in duodenum, jejunum and ileum were gradually declined in turn. Notably, the IgA+ and IgM+ cells densities change trend was the same as the total densities, and the differences among them were significant, respectively (P < 0.05). The IgG+ cells density in duodenum was the highest, followed by ileum and jejunum and there was significantly difference among them (P < 0.05). The comparison results between Control and Infected groups showed that from the duodenum, jejunum to ileum, IgA+, IgG+, and IgM+ cells were all reduced significantly, respectively. The results suggest that the three types of positive cells were resided heterogeneously in the small intestinal mucosa, that is, significant region-specificity; Moniezia benedeni infection could not change their diffuse distribution characteristics, but strikingly, reduce their resident densities, and the forming mucosal immune barrier were significantly inhibited. It provided powerful evidence for studying on the molecular mechanism of Moniezia benedeni evasion from immune surveillance by strongly inhibiting the host's mucosal immune barrier.
Collapse
|
149
|
Guan X, Zhu J, Sun H, Zhao X, Yang M, Huang Y, Pan H, Zhao Y, Zhao S. Analysis of Gut Microbiota and Metabolites in Diannan Small Ear Sows at Diestrus and Metestrus. Front Microbiol 2022; 13:826881. [PMID: 35516431 PMCID: PMC9062660 DOI: 10.3389/fmicb.2022.826881] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
The physiological state of the host affects the gut microbes. The estrus cycle is critical to the reproductive cycle of sows. However, the association between gut microbes and animal estrus is poorly understood. Here, high-throughput 16S rRNA sequencing and liquid chromatography-mass spectrometry (LC-MS) non-targeted metabolome technology were used to study the estrous cycles in Diannan small ear pigs. Significantly different gut microbiota and metabolites of sows at estrous and diestrus were screened out and the correlation was analyzed. We found that the intestinal microbial composition and microbial metabolism of Diannan small ear sows were significantly different at diestrus and metestrus. The abundances of Spirochaetes, Spirochaetia, Spirochaetales, Spirochaetaceae, Deltaproteobacteria, unidentified_Alphaproteobacteria, Ruminococcus_sp_YE281, and Treponema_berlinense in intestinal microorganisms of Diannan small ear sows at metestrus are significantly higher than that at diestrus. Propionic acid, benzyl butyrate, sucrose, piperidine, and 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) were significantly enriched at metestrus compared with diestrus, which were involved in the energy metabolism-related pathways and activated protein kinase (AMPK) signaling pathway. At diestrus and metestrus, differential microbiota of unidentified_Alphaproteobacteria, Intestinimonas, Peptococcus, Terrisporobacter, and differential metabolites of piperidine, propionic acid, and benzyl butyrate, sucrose, 4-methyl catechol, and AICAR exist a certain degree of correlation. Therefore, unidentified_Alphaproteobacteria, Ruminococcus_sp_YE281, and Treponema_berlinense may have a potential role at metestrus of the Diannan small ear sows. AICAR may be apotential marker of estrus Diannan small ear sows feces, but further studies about the specific mechanism are needed. These findings provide a new perspective for sows production management and improving sows reproductive performance.
Collapse
Affiliation(s)
- Xuancheng Guan
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
| | - Junhong Zhu
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
| | - Haichao Sun
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
| | - Xiaoqi Zhao
- Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, China
| | - Minghua Yang
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
| | - Ying Huang
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
| | - Hongbin Pan
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
| | - Yanguang Zhao
- Shanghai Laboratory Animal Research Center, Shanghai, China
| | - Sumei Zhao
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
150
|
Shi L, Yang X, Dou H, Lyu T, Wang L, Zhou S, Shang Y, Dong Y, Zhang H. Comparative Analysis of the Gut Microbiota of Mongolian Gazelle ( Procapra gutturosa) Under Fragmented Habitats. Front Microbiol 2022; 13:830321. [PMID: 35369477 PMCID: PMC8965509 DOI: 10.3389/fmicb.2022.830321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/11/2022] [Indexed: 12/15/2022] Open
Abstract
The existence of man-made facilities such as pasture fences makes the grassland ecosystem fragmented and endangers the survival of local wild animals. The Mongolian gazelle is highly sensitive to hunting and habitat destruction, and is one of the most threatened artiodactyls in Eurasia. It provides a critical model to studying gut microbiota under fragmented habitats. Therefore, we applied metagenomics sequencing to analyze the gut microbiota communities and functions of Mongolian gazelle under fragmented habitats. The results demonstrated that there were no significant differences in gut microbial communities between the different groups at both the phylum and genus level. The functional analyses showed that the Mongolian gazelle in fragmented habitat had a stronger ability to degrade naphthalene, but their ability to absorb carbohydrates was weaker. This study provided fundamental information about the gut microbiota of Mongolian gazelle, and we recommend reducing habitat fragmentation to better protect the Mongolian gazelle.
Collapse
Affiliation(s)
- Lupeng Shi
- College of Life Sciences, Qufu Normal University, Qufu, China
| | - Xiufeng Yang
- College of Life Sciences, Qufu Normal University, Qufu, China
| | - Huashan Dou
- Hulunbuir Academy of Inland Lakes in Northern Cold & Arid Areas, Hulunbuir, China
| | - Tianshu Lyu
- College of Life Sciences, Qufu Normal University, Qufu, China
| | - Lidong Wang
- College of Life Sciences, Qufu Normal University, Qufu, China
| | - Shengyang Zhou
- College of Life Sciences, Qufu Normal University, Qufu, China
| | - Yongquan Shang
- College of Life Sciences, Qufu Normal University, Qufu, China
| | - Yuehuan Dong
- College of Life Sciences, Qufu Normal University, Qufu, China
| | - Honghai Zhang
- College of Life Sciences, Qufu Normal University, Qufu, China
| |
Collapse
|