1
|
Li J, Jiang N, Zheng H, Zheng X, Xu Y, Weng Y, Jiang F, Wang C, Chang P. Investigation of gut microbiota disorders in norovirus infected children patients based on 16s rRNA sequencing. Ann Med 2024; 56:2412834. [PMID: 39387550 PMCID: PMC11469441 DOI: 10.1080/07853890.2024.2412834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 01/18/2024] [Accepted: 05/27/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Norovirus is the leading cause of sporadic viral gastroenteritis cases and outbreaks. Gut microbiota plays a key role in maintaining immune homeostasis. We aimed to investigate the composition and functional effects of gut microbiota in children infected with norovirus. METHODS Stool samples were collected from 31 children infected with norovirus and 25 healthy children. The gut microbiota was analyzed by 16S rRNA gene sequencing, followed by composition, correlation network, functional and phenotype prediction analyses. RESULTS Gut microbiota in children infected with norovirus was characterized by lower species richness and diversity. Veillonella is the dominant gut microbiota specie in norovirus infection. Blautia was significantly lower in norovirus infection. There was a positive correlation between Faecalibacterium, Blautia, Subdoligranulum, Eubacterium_hallii_group, Fusicatenibacter, Agathobacter, Roseburia and Dorea. Functionally, secondary metabolites biosynthesis, transport and catabolism, selenocysteine lyase and peroxiredoxin were the most significantly higher functional compositions of gut microbiota in norovirus infection. However, sn-glycerol-1-phosphate dehydrogenase and fermentation were the most significantly lower functional compositions in norovirus infection group. Phenotype analysis showed that Contains_Mobile_Elements had the highest level of phenotypes in the gut microbiota of norovirus infection. CONCLUSION Norovirus infection may lead to dysregulation of the gut microbiome in children.
Collapse
Affiliation(s)
- Jie Li
- Comprehensive Technical Service Center, Taizhou Customs, Taizhou City, Zhejiang, China
| | - Nan Jiang
- Department of Integrated Traditional Chinese and Western Medicine, Taizhou Municipal Hospital, Taizhou City, Zhejiang, China
| | - Hui Zheng
- Medical and Chemical Testing Center, Taizhou Institute of Measurement Technology, Taizhou City, Zhejiang, China
| | - Xiao Zheng
- School Office, Dongshan Central Primary School, Taizhou City, Zhejiang, China
| | - Yi Xu
- Comprehensive Technical Service Center, Taizhou Customs, Taizhou City, Zhejiang, China
| | - Yongqing Weng
- Clinical Laboratory, Taizhou Municipal Hospital, Taizhou City, Zhejiang, China
| | - Feijian Jiang
- Supervision Section No. 1, Taizhou Customs, Taizhou City, Zhejiang, China
| | - Chong Wang
- Inspection Section, Taizhou Customs, Taizhou City, Zhejiang, China
| | - Peiliang Chang
- Supervision Section No. 4, Taizhou Customs, Taizhou City, Zhejiang, China
| |
Collapse
|
2
|
Tesfaw G, Siraj DS, Abdissa A, Jakobsen RR, Johansen ØH, Zangenberg M, Hanevik K, Mekonnen Z, Langeland N, Bjørang O, Safdar N, Mapes AC, Kates A, Krych L, Castro-Mejía JL, Nielsen DS. Gut microbiota patterns associated with duration of diarrhea in children under five years of age in Ethiopia. Nat Commun 2024; 15:7532. [PMID: 39223134 PMCID: PMC11369280 DOI: 10.1038/s41467-024-51464-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Diarrhea claims >500,000 lives annually among children under five years of age in low- and middle-income countries. Mortality due to acute diarrhea (<7 days' duration) is decreasing, but prolonged (7-13 days) and persistent (≥14 days of duration) diarrhea remains a massive challenge. Here, we use a case-control study to decipher if fecal gut microbiota compositional differences between Ethiopian children with acute (n=554) or prolonged/persistent (n=95) diarrhea and frequency-matched non-diarrheal controls (n=663) are linked to diarrheal etiology. We show that diarrhea cases are associated with lower bacterial diversity and enriched in Escherichia spp., Campylobacter spp., and Streptococcus spp. Further, diarrhea cases are depleted in gut commensals such as Prevotella copri, Faecalibacterium prausnitzii, and Dialister succinatiphilus, with depletion being most pronounced in prolonged/persistent cases, suggesting that prolonged duration of diarrhea is accompanied by depletion of gut commensals and that re-establishing these via e.g., microbiota-directed food supplements offer a potential treatment strategy.
Collapse
Affiliation(s)
- Getnet Tesfaw
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark.
- School of Medical Laboratory Sciences, Jimma University, Jimma, Ethiopia.
| | - Dawd S Siraj
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Alemseged Abdissa
- School of Medical Laboratory Sciences, Jimma University, Jimma, Ethiopia
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | | | - Øystein H Johansen
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Microbiology, Vestfold Hospital Trust, Tønsberg, Norway
- Microbiology Laboratory, Southern Health and Social Care Trust, Portadown, Northern Ireland
| | - Mike Zangenberg
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark
| | - Kurt Hanevik
- Department of Clinical Science, University of Bergen, Bergen, Norway
- National Center for Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Zeleke Mekonnen
- School of Medical Laboratory Sciences, Jimma University, Jimma, Ethiopia
| | - Nina Langeland
- Department of Clinical Science, University of Bergen, Bergen, Norway
- National Center for Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Ola Bjørang
- Department of Microbiology, Vestfold Hospital Trust, Tønsberg, Norway
| | - Nasia Safdar
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Abigail C Mapes
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Ashley Kates
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Lukasz Krych
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | | | - Dennis S Nielsen
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
3
|
Kim YI, Kim SY, Lee S, Kim M, Kim WJ. Investigation of gut microbiota diversity according to infectious agent in pediatric infectious acute gastroenteritis in a Korean university hospital. Pediatr Neonatol 2024; 65:476-481. [PMID: 38471992 DOI: 10.1016/j.pedneo.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 12/19/2023] [Accepted: 01/26/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Acute gastroenteritis (AGE) is a common cause of pediatric morbidity and mortality worldwide. AGE can cause an imbalance in the intestinal microbiota. This study aimed to investigate the diversity of the gut microbiome in Korean children hospitalized for infectious AGE at a university hospital. METHODS A total of 23 stool samples from patients aged 5 months to 11 years with AGE were analyzed. Thirteen convalescent stool samples were collected 1 month after discharge. Multiplex polymerase chain reaction (PCR) for the five viruses and 16 bacteria-specific AGE pathogens (PowerChek Multiplex Real time PCR Kit, Seoul, Korea), and 16s rRNA sequencing (Illumina MiSeq Sequencing system, Illumina, USA) were performed. RESULTS According to the results of multiplex PCR for causative pathogens, the microbiome taxonomic profile (MTP) of the gut microbiome in three groups of AGE, norovirus AGE (n = 11), Campylobacter AGE (n = 7) and Salmonella AGE (n = 5) was compared. The phylum Actinobacteria was significantly more abundant in the norovirus AGE (P = 0.011), whereas the phylum Proteobacteria was significantly more abundant in Salmonella AGE (P = 0.012). Alpha diversity, which indicates species richness and diversity, showed no statistical differences. However, beta diversity, representing the similarity in MTP between norovirus, Campylobacter, and Salmonella AGE, was significantly different (P = 0.007). In convalescence, compared with their corresponding AGE samples, the phylum Firmicutes; and the lower taxa Christensenellaceae (P = 0.0152) and Lachnospiraceae (P = 0.0327) were significantly increased. CONCLUSIONS In pediatric AGE, the type of infectious agent can affect the diversity and dominance of gut microbiota in pediatric patients. Furthermore, healthy gut bacteria increased during the period of 1 month after infection, allowing a return to a healthy state without causing long-term dysbiosis.
Collapse
Affiliation(s)
- You Ie Kim
- Department of Pediatrics, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang Yong Kim
- Department of Pediatrics, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seungok Lee
- Department of Laboratory Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Myungshin Kim
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Woo Jin Kim
- EONE Laboratories, Incheon, Republic of Korea
| |
Collapse
|
4
|
Lee J, Hunter B, Shim H. A pangenome analysis of ESKAPE bacteriophages: the underrepresentation may impact machine learning models. Front Mol Biosci 2024; 11:1395450. [PMID: 38974320 PMCID: PMC11224154 DOI: 10.3389/fmolb.2024.1395450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/31/2024] [Indexed: 07/09/2024] Open
Abstract
Bacteriophages are the most prevalent biological entities in the biosphere. However, limitations in both medical relevance and sequencing technologies have led to a systematic underestimation of the genetic diversity within phages. This underrepresentation not only creates a significant gap in our understanding of phage roles across diverse biosystems but also introduces biases in computational models reliant on these data for training and testing. In this study, we focused on publicly available genomes of bacteriophages infecting high-priority ESKAPE pathogens to show the extent and impact of this underrepresentation. First, we demonstrate a stark underrepresentation of ESKAPE phage genomes within the public genome and protein databases. Next, a pangenome analysis of these ESKAPE phages reveals extensive sharing of core genes among phages infecting the same host. Furthermore, genome analyses and clustering highlight close nucleotide-level relationships among the ESKAPE phages, raising concerns about the limited diversity within current public databases. Lastly, we uncover a scarcity of unique lytic phages and phage proteins with antimicrobial activities against ESKAPE pathogens. This comprehensive analysis of the ESKAPE phages underscores the severity of underrepresentation and its potential implications. This lack of diversity in phage genomes may restrict the resurgence of phage therapy and cause biased outcomes in data-driven computational models due to incomplete and unbalanced biological datasets.
Collapse
Affiliation(s)
- Jeesu Lee
- Center for Biosystems and Biotech Data Science, Ghent University Global Campus, Incheon, Republic of Korea
| | - Branden Hunter
- Department of Biology, California State University, Fresno, CA, United States
| | - Hyunjin Shim
- Center for Biosystems and Biotech Data Science, Ghent University Global Campus, Incheon, Republic of Korea
- Department of Biology, California State University, Fresno, CA, United States
| |
Collapse
|
5
|
Rimkute I, Chaimongkol N, Woods KD, Nagata BM, Darko S, Gudbole S, Henry AR, Sosnovtsev SV, Olia AS, Verardi R, Bok K, Todd JP, Woodward R, Kwong PD, Douek DC, Alves DA, Green KY, Roederer M. A non-human primate model for human norovirus infection. Nat Microbiol 2024; 9:776-786. [PMID: 38321182 DOI: 10.1038/s41564-023-01585-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/12/2023] [Indexed: 02/08/2024]
Abstract
Norovirus infection can cause gastrointestinal disease in humans. Development of therapies and vaccines against norovirus have been limited by the lack of a suitable and reliable animal model. Here we established rhesus macaques as an animal model for human norovirus infection. We show that rhesus macaques are susceptible to oral infection with human noroviruses from two different genogroups. Variation in duration of virus shedding (days to weeks) between animals, evolution of the virus over the time of infection, induction of virus-specific adaptive immune responses, susceptibility to reinfection and preferential replication of norovirus in the jejunum of rhesus macaques was similar to infection reported in humans. We found minor pathological signs and changes in epithelial cell surface glycosylation patterns in the small intestine during infection. Detection of viral protein and RNA in intestinal biopsies confirmed the presence of the virus in chromogranin A-expressing epithelial cells, as it does in humans. Thus, rhesus macaques are a promising non-human primate model to evaluate vaccines and therapeutics against norovirus disease.
Collapse
Affiliation(s)
- Inga Rimkute
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Natthawan Chaimongkol
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kamron D Woods
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Bianca M Nagata
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| | - Samuel Darko
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Sucheta Gudbole
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Amy R Henry
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Stanislav V Sosnovtsev
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Adam S Olia
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Raffaello Verardi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Karin Bok
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - John-Paul Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Ruth Woodward
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Derron A Alves
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| | - Kim Y Green
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
6
|
Montalvo-Otivo R, Vilcapoma P, Murillo A, Mathey C, Olivera A, Veliz G, Estrella D. Evaluation of chronic diarrhea in patients newly diagnosed with HIV infection through the FilmArray® gastrointestinal panel. REVISTA DE GASTROENTEROLOGIA DE MEXICO (ENGLISH) 2024; 89:80-88. [PMID: 36890062 DOI: 10.1016/j.rgmxen.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 11/16/2022] [Indexed: 03/08/2023]
Abstract
INTRODUCTION The treatment and diagnosis of chronic diarrhea in the immunocompromised patient depends on the ability to rapidly detect the etiologic agents. AIMS Our aim was to evaluate the results of the FilmArray® gastrointestinal panel in patients newly diagnosed with HIV infection that presented with chronic diarrhea. MATERIAL AND METHODS Utilizing nonprobability consecutive convenience sampling, 24 patients were included that underwent molecular testing for the simultaneous detection of 22 pathogens. RESULTS In 24 HIV-infected patients with chronic diarrhea, enteropathogen bacteria were detected in 69% of the cases, parasites in 18%, and viruses in 13%. Enteropathogenic Escherichia coli and enteroaggregative Escherichia coli were the main bacteria identified, Giardia lamblia was found in 25%, and norovirus was the most frequent viral agent. The median number of infectious agents per patient was three (range of 0 to 7). The biologic agents not identified through the FilmArray® method were tuberculosis and fungi. CONCLUSIONS Several infectious agents were simultaneously detected through the FilmArray® gastrointestinal panel in patients with HIV infection and chronic diarrhea.
Collapse
Affiliation(s)
| | - P Vilcapoma
- Facultad de Medicina Humana, Universidad Continental, Huancayo, Peru
| | - A Murillo
- Servicio de Enfermedades Infecciosas, Hospital Daniel Alcides Carrión, Huancayo, Peru
| | - C Mathey
- Facultad de Medicina Humana, Universidad Continental, Huancayo, Peru
| | - A Olivera
- Facultad de Medicina Humana, Universidad Peruana Los Andes, Huancayo, Peru
| | - G Veliz
- Facultad de Medicina Humana, Universidad Peruana Los Andes, Huancayo, Peru
| | - D Estrella
- Servicio de Microbiología, Hospital Daniel Alcides Carrión, Huancayo, Peru
| |
Collapse
|
7
|
Cupido DT, de Beer C. Screening for viral pathogens in the gastrointestinal tract from cases of sudden unexpected death in infancy at the Tygerberg Medico-legal Mortuary. Virol J 2023; 20:281. [PMID: 38031160 PMCID: PMC10688011 DOI: 10.1186/s12985-023-02249-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/19/2023] [Indexed: 12/01/2023] Open
Abstract
Sudden and unexpected death in infancy (SUDI) may be triggered by an external risk or exposure. Intestinal infections with enteric viruses may disrupt the gut and enhance bacterial toxins present in SUDI cases. While diarrhoeal disease deaths have decreased worldwide, approximately half a million deaths still occur in children in Sub- Saharan Africa and South Asia. Furthermore, the role of viral enteropathogens in SUDI cases have not been investigated. The aim of this study was to describe specific viral pathogens in stool samples collected from SUDI cases and age-matched, apparently healthy infants in Cape Town, South Africa. Stool samples were collected from 176 SUDI cases between June 2017 and May 2018. In addition, stool samples were collected from the nappies of 30 age-matched, apparently healthy infants as a control group. Real-time polymerase chain reaction was performed on the stool samples for viral detection. A total of 111 SUDI cases were positive for viruses, with rotavirus (38.6%; 68/176) and norovirus GI and GII (30.0%; 53/176) were prevalent in SUDI cases. Adenovirus Type F was present in only 15.9% (28/176), astrovirus in 9.7% (17/176), and sapovirus in 0.6% (1/176) of cases. In the control samples, norovirus GII was detected most frequently (36.7%; 11/30), followed by rotavirus (33.3%; 10/30), and sapovirus in 6.7% (2/30). While there was no significant association between SUDI cases and enteric viruses, the majority of viruses were significantly associated with the seasons. The study confirms the importance of rotavirus vaccination and describes the significance of norovirus infection in children, post rotavirus vaccine introduction.
Collapse
Affiliation(s)
- Danielle T Cupido
- Division of Medical Virology, Department of Pathology, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa.
| | - Corena de Beer
- Division of Medical Virology, Department of Pathology, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa.
| |
Collapse
|
8
|
Armah G, Lopman BA, Vinjé J, O'Ryan M, Lanata CF, Groome M, Ovitt J, Marshall C, Sajewski E, Riddle MS. Vaccine value profile for norovirus. Vaccine 2023; 41 Suppl 2:S134-S152. [PMID: 37951692 PMCID: PMC10710898 DOI: 10.1016/j.vaccine.2023.03.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/11/2023] [Accepted: 03/16/2023] [Indexed: 11/14/2023]
Abstract
Norovirus is attributed to nearly 1 out of every 5 episodes of diarrheal disease globally and is estimated to cause approximately 200,000 deaths annually worldwide, with 70,000 or more among children in developing countries. Noroviruses remain a leading cause of sporadic disease and outbreaks of acute gastroenteritis even in industrialized settings, highlighting that improved hygiene and sanitation alone may not be fully effective in controlling norovirus. Strengths in global progress towards a Norovirus vaccine include a diverse though not deep pipeline which includes multiple approaches, including some with proven technology platforms (e.g., VLP-based HPV vaccines). However, several gaps in knowledge persist, including a fulsome mechanistic understanding of how the virus attaches to human host cells, internalizes, and induces disease.
Collapse
Affiliation(s)
- George Armah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Ben A Lopman
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Jan Vinjé
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Miguel O'Ryan
- Microbiology and Mycology Program, Faculty of Medicine, University of Chile and Instituto de Sistemas Complejos de Ingenierìa (ISCI), Santiago, Chile
| | | | - Michelle Groome
- National Institute for Communicable Diseases, National Health Laboratory Services, Johannesburg, South Africa; School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jared Ovitt
- Office of Medical Research, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | | | - Elizabeth Sajewski
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Mark S Riddle
- Office of Medical Research, University of Nevada, Reno School of Medicine, Reno, Nevada, USA.
| |
Collapse
|
9
|
Carreno-Florez GP, Kocak BR, Hendricks MR, Melvin JA, Mar KB, Kosanovich J, Cumberland RL, Delgoffe GM, Shiva S, Empey KM, Schoggins JW, Bomberger JM. Interferon signaling drives epithelial metabolic reprogramming to promote secondary bacterial infection. PLoS Pathog 2023; 19:e1011719. [PMID: 37939149 PMCID: PMC10631704 DOI: 10.1371/journal.ppat.1011719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/28/2023] [Indexed: 11/10/2023] Open
Abstract
Clinical studies report that viral infections promote acute or chronic bacterial infections at multiple host sites. These viral-bacterial co-infections are widely linked to more severe clinical outcomes. In experimental models in vitro and in vivo, virus-induced interferon responses can augment host susceptibility to secondary bacterial infection. Here, we used a cell-based screen to assess 389 interferon-stimulated genes (ISGs) for their ability to induce chronic Pseudomonas aeruginosa infection. We identified and validated five ISGs that were sufficient to promote bacterial infection. Furthermore, we dissected the mechanism of action of hexokinase 2 (HK2), a gene involved in the induction of aerobic glycolysis, commonly known as the Warburg effect. We report that HK2 upregulation mediates the induction of Warburg effect and secretion of L-lactate, which enhances chronic P. aeruginosa infection. These findings elucidate how the antiviral immune response renders the host susceptible to secondary bacterial infection, revealing potential strategies for viral-bacterial co-infection treatment.
Collapse
Affiliation(s)
- Grace P. Carreno-Florez
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Brian R. Kocak
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Matthew R. Hendricks
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jeffrey A. Melvin
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Katrina B. Mar
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jessica Kosanovich
- Department of Pharmacy and Therapeutics and Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, United States of America
| | - Rachel L. Cumberland
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Greg M. Delgoffe
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Kerry M. Empey
- Department of Pharmacy and Therapeutics and Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - John W. Schoggins
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jennifer M. Bomberger
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| |
Collapse
|
10
|
Gupta U, Dey P. Rise of the guardians: Gut microbial maneuvers in bacterial infections. Life Sci 2023; 330:121993. [PMID: 37536616 DOI: 10.1016/j.lfs.2023.121993] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/23/2023] [Accepted: 07/29/2023] [Indexed: 08/05/2023]
Abstract
AIMS Bacterial infections are one of the major causes of mortality globally. The gut microbiota, primarily comprised of the commensals, performs an important role in maintaining intestinal immunometabolic homeostasis. The current review aims to provide a comprehensive understanding of how modulation of the gut microbiota influences opportunistic bacterial infections. MATERIALS AND METHODS Primarily centered around mechanisms related to colonization resistance, nutrient, and metabolite-associated factors, mucosal immune response, and commensal-pathogen reciprocal interactions, we discuss how gut microbiota can promote or prevent bacterial infections. KEY FINDINGS Opportunistic infections can occur directly due to obligate pathogens or indirectly due to the overgrowth of opportunistic pathobionts. Gut microbiota-centered mechanisms of altered intestinal immunometabolic and metabolomic homeostasis play a significant role in infection promotion and prevention. Depletion in the population of commensals, increased abundance of pathobionts, and overall decrease in gut microbial diversity and richness caused due to prolonged antibiotic use are risk factors of opportunistic bacterial infections, including infections from multidrug-resistant spp. Gut commensals can limit opportunistic infections by mechanisms including the production of antimicrobials, short-chain fatty acids, bile acid metabolism, promoting mucin formation, and maintaining immunological balance at the mucosa. Gut microbiota-centered strategies, including the administration of probiotics and fecal microbiota transplantation, could help attenuate opportunistic bacterial infections. SIGNIFICANCE The current review discussed the gut microbial population and function-specific aspects contributing to bacterial infection susceptibility and prophylaxis. Collectively, this review provides a comprehensive understanding of the mechanisms related to the dual role of gut microbiota in bacterial infections.
Collapse
Affiliation(s)
- Upasana Gupta
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, Punjab, India
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, Punjab, India.
| |
Collapse
|
11
|
Abstract
The mammalian gastrointestinal tract (GIT) hosts a diverse and highly active microbiota composed of bacteria, eukaryotes, archaea, and viruses. Studies of the GIT microbiota date back more than a century, although modern techniques, including mouse models, sequencing technology, and novel therapeutics in humans, have been foundational to our understanding of the roles of commensal microbes in health and disease. Here, we review the impacts of the GIT microbiota on viral infection, both within the GIT and systemically. GIT-associated microbes and their metabolites alter the course of viral infection through a variety of mechanisms, including direct interactions with virions, alteration of the GIT landscape, and extensive regulation of innate and adaptive immunity. Mechanistic understanding of the full breadth of interactions between the GIT microbiota and the host is still lacking in many ways but will be vital for the development of novel therapeutics for viral and nonviral diseases alike.
Collapse
Affiliation(s)
- Danielle E Campbell
- Department of Medicine, Division of Infectious Diseases and Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Yuhao Li
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Harshad Ingle
- Department of Medicine, Division of Infectious Diseases and Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Megan T Baldridge
- Department of Medicine, Division of Infectious Diseases and Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA;
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
12
|
Xiao Q, Chen B, Zhu Z, Yang T, Tao E, Hu C, Zheng W, Tang W, Shu X, Jiang M. Alterations in the Fecal Microbiota Composition in Pediatric Acute Diarrhea: A Cross-Sectional and Comparative Study of Viral and Bacterial Enteritis. Infect Drug Resist 2023; 16:5473-5483. [PMID: 37638073 PMCID: PMC10456034 DOI: 10.2147/idr.s410720] [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: 03/23/2023] [Accepted: 08/12/2023] [Indexed: 08/29/2023] Open
Abstract
Objective To examine the association between the fecal microbiota of acute diarrhea in children and provide gut microbiota information related the acute diarrhea with rotavirus. Patients and Methods Children with acute diarrhea aged 3-60 months were selected for the study. Routine stool examination was performed, and stool samples were collected and stored at -80 °C until further analysis. Fecal microbial DNA was extracted, and DNA concentration and quality were detected. PCR amplification and 16S rDNA high-throughput sequencing analysis using the Illumina MiSeq platform were performed, and intestinal flora was statistically analyzed. Results Children with acute diarrhea exhibited gut microbial dysbiosis. Lower microbial diversity and richness were observed in the viral enteritis and bacterial enteritis groups than in the control group. Composition of the microbiota in acute diarrhea differed from that in the control group. The Bacteroidetes/Firmicutes dramatically decreased in the viral enteritis and bacterial enteritis groups. However, the relative abundance of Proteobacteria and Fusobacteria increased, especially in the bacterial enteritis group. In addition, the relative abundance of Actinobacteria had dramatically increased in the viral enteritis group. According to the Kyoto Encyclopedia of Genes and Genomes map analysis, the membrane transport dysfunction was caused by rotavirus infection, while the membrane transport dysfunction was more evident in bacterial infection. Conclusion Acute diarrhea infections cause fecal microbiota dysbiosis in children. Changes in fecal microflora in children suggest that the regulation of intestinal flora in children with acute diarrhea should be strengthened.
Collapse
Affiliation(s)
- Qiulin Xiao
- Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
- Department of Gastroenterology, Hangzhou Children’s Hospital, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Bo Chen
- Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
| | - Zhenya Zhu
- Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
| | - Ting Yang
- Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
| | - Enfu Tao
- Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
| | - Chenmin Hu
- Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
| | - Wei Zheng
- Department of Gastroenterology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
| | - Weihong Tang
- Department of Gastroenterology, Hangzhou Children’s Hospital, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Xiaoli Shu
- Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
| | - Mizu Jiang
- Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
- Department of Gastroenterology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
| |
Collapse
|
13
|
Quaye EK, Adjei RL, Isawumi A, Allen DJ, Caporaso JG, Quaye O. Altered Faecal Microbiota Composition and Structure of Ghanaian Children with Acute Gastroenteritis. Int J Mol Sci 2023; 24:3607. [PMID: 36835017 PMCID: PMC9962333 DOI: 10.3390/ijms24043607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 02/15/2023] Open
Abstract
Acute gastroenteritis (AGE) is a disease of global public health importance. Recent studies show that children with AGE have an altered gut microbiota relative to non-AGE controls. Yet, how the gut microbiota differs in Ghanaian children with and without AGE remains unclear. Here, we explore the 16S rRNA gene-based faecal microbiota profiles of Ghanaian children five years of age and younger, comprising 57 AGE cases and 50 healthy controls. We found that AGE cases were associated with lower microbial diversity and altered microbial sequence profiles relative to the controls. The faecal microbiota of AGE cases was enriched for disease-associated bacterial genera, including Enterococcus, Streptococcus, and Staphylococcus. In contrast, the faecal microbiota of controls was enriched for potentially beneficial genera, including Faecalibacterium, Prevotella, Ruminococcus, and Bacteroides. Lastly, distinct microbial correlation network characteristics were observed between AGE cases and controls, thereby supporting broad differences in faecal microbiota structure. Altogether, we show that the faecal microbiota of Ghanaian children with AGE differ from controls and are enriched for bacterial genera increasingly associated with diseases.
Collapse
Affiliation(s)
- Emmanuel Kofi Quaye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| | - Raymond Lovelace Adjei
- Council for Scientific and Industrial Research (CSIR)-Animal Research Institute, Accra P.O. Box AH 20, Ghana
| | - Abiola Isawumi
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| | - David J. Allen
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
- Vaccine Centre, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - J. Gregory Caporaso
- Centre for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ 86011, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Osbourne Quaye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| |
Collapse
|
14
|
Montalvo-Otivo R, Vilcapoma P, Murillo A, Mathey C, Olivera A, Veliz G, Estrella D. Evaluación de la diarrea crónica en pacientes con reciente diagnóstico de infección por VIH mediante el panel gastrointestinal FilmArray®. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO 2023. [DOI: 10.1016/j.rgmx.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
|
15
|
Miliwebsky E, Jure MÁ, Farfan MJ, Palermo MS. Interactions of Pathogenic Escherichia coli with Gut Microbiota. TRENDING TOPICS IN ESCHERICHIA COLI RESEARCH 2023:277-294. [DOI: 10.1007/978-3-031-29882-0_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
16
|
Pearce VH, Groisman EA, Townsend GE. Dietary sugars silence the master regulator of carbohydrate utilization in human gut Bacteroides species. Gut Microbes 2023; 15:2221484. [PMID: 37358144 PMCID: PMC10294740 DOI: 10.1080/19490976.2023.2221484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/08/2023] [Indexed: 06/27/2023] Open
Abstract
The mammalian gut microbiota is a critical human health determinant with therapeutic potential for remediation of many diseases. The host diet is a key factor governing the gut microbiota composition by altering nutrient availability and supporting the expansion of distinct microbial populations. Diets rich in simple sugars modify the abundance of microbial subsets, enriching for microbiotas that elicit pathogenic outcomes. We previously demonstrated that diets rich in fructose and glucose can reduce the fitness and abundance of a human gut symbiont, Bacteroides thetaiotaomicron, by silencing the production of a critical intestinal colonization protein, called Roc, via its mRNA leader through an unknown mechanism. We have now determined that dietary sugars silence Roc by reducing the activity of BT4338, a master regulator of carbohydrate utilization. Here, we demonstrate that BT4338 is required for Roc synthesis, and that BT4338 activity is silenced by glucose or fructose. We show that the consequences of glucose and fructose on orthologous transcription factors are conserved across human intestinal Bacteroides species. This work identifies a molecular pathway by which a common dietary additive alters microbial gene expression in the gut that could be harnessed to modulate targeted microbial populations for future therapeutic interventions.
Collapse
Affiliation(s)
- Victoria H. Pearce
- Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA, USA
- Penn State Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| | - Eduardo A. Groisman
- Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
- Microbial Sciences Institute, Yale University, New Haven, CT, USA
| | - Guy E. Townsend
- Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA, USA
- Penn State Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| |
Collapse
|
17
|
Spivak NM, Haroon J, Swenson A, Turnbull SA, Dang N, Ganeles M, Price C, Distler M, Nurmi E, Lavretsky H, Bystritsky A. Microbiome in Anxiety and Other Psychiatric Disorders. Med Clin North Am 2023; 107:73-83. [PMID: 36402501 DOI: 10.1016/j.mcna.2022.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Initial studies suggested that the fluctuations in the quantity, variety, and composition of the gut microbiota can significantly affect disease processes. This change in the gut microbiota causing negative health benefits was coined dysbiosis. Initial research focused on gastrointestinal illnesses. However, the gut microbiome was found to affect more than just gastrointestinal diseases. Numerous studies have proven that the gut microbiome can influence neuropsychiatric diseases such as Parkinson's disease, Alzheimer's disease, and multiple sclerosis.
Collapse
Affiliation(s)
- Norman M Spivak
- UCLA-Caltech Medical Scientist Training Program; Department of Psychiatry and Biobehavioral Sciences, DGSOM, UCLA, 300 UCLA Medical Plaza, Suite 2200, Los Angeles, CA 90095, USA.
| | - Jonathan Haroon
- Department of Psychiatry and Biobehavioral Sciences, DGSOM, UCLA, 300 UCLA Medical Plaza, Suite 2200, Los Angeles, CA 90095, USA
| | - Andrew Swenson
- Department of Psychiatry and Biobehavioral Sciences, DGSOM, UCLA, 300 UCLA Medical Plaza, Suite 2200, Los Angeles, CA 90095, USA
| | - Scott A Turnbull
- Department of Internal Medicine, Kirk Kerkorian SOM, UNLV, 4505 South Maryland Parkway, Las Vegas, NV 89154, USA
| | - Nolan Dang
- Department of Psychiatry and Biobehavioral Sciences, DGSOM, UCLA, 300 UCLA Medical Plaza, Suite 2200, Los Angeles, CA 90095, USA
| | - Matthew Ganeles
- Department of Molecular, Cell and Developmental Biology, UCLA, 300 UCLA Medical Plaza, Suite 2200, Los Angeles, CA 90095, USA
| | - Collin Price
- Department of Psychiatry and Biobehavioral Sciences, DGSOM, UCLA, 300 UCLA Medical Plaza, Suite 2200, Los Angeles, CA 90095, USA
| | - Margaret Distler
- Department of Psychiatry and Biobehavioral Sciences, DGSOM, UCLA, 300 UCLA Medical Plaza, Suite 2200, Los Angeles, CA 90095, USA
| | - Erika Nurmi
- Department of Psychiatry and Biobehavioral Sciences, DGSOM, UCLA, 300 UCLA Medical Plaza, Suite 2200, Los Angeles, CA 90095, USA
| | - Helen Lavretsky
- Department of Psychiatry and Biobehavioral Sciences, DGSOM, UCLA, 300 UCLA Medical Plaza, Suite 2200, Los Angeles, CA 90095, USA
| | - Alexander Bystritsky
- Department of Psychiatry and Biobehavioral Sciences, DGSOM, UCLA, 300 UCLA Medical Plaza, Suite 2200, Los Angeles, CA 90095, USA
| |
Collapse
|
18
|
Schmid DW, Fackelmann G, Wasimuddin, Rakotondranary J, Ratovonamana YR, Montero BK, Ganzhorn JU, Sommer S. A framework for testing the impact of co-infections on host gut microbiomes. Anim Microbiome 2022; 4:48. [PMID: 35945629 PMCID: PMC9361228 DOI: 10.1186/s42523-022-00198-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 07/26/2022] [Indexed: 02/07/2023] Open
Abstract
Parasitic infections disturb gut microbial communities beyond their natural range of variation, possibly leading to dysbiosis. Yet it remains underappreciated that most infections are accompanied by one or more co-infections and their collective impact is largely unexplored. Here we developed a framework illustrating changes to the host gut microbiome following single infections, and build on it by describing the neutral, synergistic or antagonistic impacts on microbial α- and ß-diversity expected from co-infections. We tested the framework on microbiome data from a non-human primate population co-infected with helminths and Adenovirus, and matched patterns reported in published studies to the introduced framework. In this case study, α-diversity of co-infected Malagasy mouse lemurs (Microcebus griseorufus) did not differ in comparison with that of singly infected or uninfected individuals, even though community composition captured with ß-diversity metrices changed significantly. Explicitly, we record stochastic changes in dispersion, a sign of dysbiosis, following the Anna-Karenina principle rather than deterministic shifts in the microbial gut community. From the literature review and our case study, neutral and synergistic impacts emerged as common outcomes from co-infections, wherein both shifts and dispersion of microbial communities following co-infections were often more severe than after a single infection alone, but microbial α-diversity was not universally altered. Important functions of the microbiome may also suffer from such heavily altered, though no less species-rich microbial community. Lastly, we pose the hypothesis that the reshuffling of host-associated microbial communities due to the impact of various, often coinciding parasitic infections may become a source of novel or zoonotic diseases.
Collapse
|
19
|
Wahab S, Almaghaslah D, Mahmood SE, Ahmad MF, Alsayegh AA, Abu Haddash YM, Rahman MA, Ahamd I, Ahmad W, Khalid M, Usmani S, Ahmad MP, Hani U. Pharmacological Efficacy of Probiotics in Respiratory Viral Infections: A Comprehensive Review. J Pers Med 2022; 12:1292. [PMID: 36013241 PMCID: PMC9409792 DOI: 10.3390/jpm12081292] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 01/30/2023] Open
Abstract
Mortality and morbidity from influenza and other respiratory viruses are significant causes of concern worldwide. Infections in the respiratory tract are often underappreciated because they tend to be mild and incapacitated. On the other hand, these infections are regarded as a common concern in clinical practice. Antibiotics are used to treat bacterial infections, albeit this is becoming more challenging since many of the more prevalent infection causes have acquired a wide range of antimicrobial resistance. Resistance to frontline treatment medications is constantly rising, necessitating the development of new antiviral agents. Probiotics are one of several medications explored to treat respiratory viral infection (RVI). As a result, certain probiotics effectively prevent gastrointestinal dysbiosis and decrease the likelihood of secondary infections. Various probiotic bacterias and their metabolites have shown immunomodulating and antiviral properties. Unfortunately, the mechanisms by which probiotics are effective in the fight against viral infections are sometimes unclear. This comprehensive review has addressed probiotic strains, dosage regimens, production procedures, delivery systems, and pre-clinical and clinical research. In particular, novel probiotics' fight against RVIs is the impetus for this study. Finally, this review may explore the potential of probiotic bacterias and their metabolites to treat RVIs. It is expected that probiotic-based antiviral research would be benefitted from this review's findings.
Collapse
Affiliation(s)
- Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Dalia Almaghaslah
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Syed Esam Mahmood
- Department of Family and Community Medicine, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - Md Faruque Ahmad
- Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Abdulrahman A. Alsayegh
- Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Yahya M. Abu Haddash
- Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Mohammad Akhlaquer Rahman
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, Taif 21974, Saudi Arabia
| | - Irfan Ahamd
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Wasim Ahmad
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Safaa, Dammam 34222, Saudi Arabia
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Shazia Usmani
- Herbal Bioactive Research Laboratory, Faculty of Pharmacy, Integral University, Dasauli, Kursi Road, Lucknow 226026, Uttar Pradesh, India
| | - Md Parwez Ahmad
- Department of Pharmacology, School of Medicine, Maldives National University, Male 20402, Maldives
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| |
Collapse
|
20
|
Gough EK. The impact of mass drug administration of antibiotics on the gut microbiota of target populations. Infect Dis Poverty 2022; 11:76. [PMID: 35773678 PMCID: PMC9245274 DOI: 10.1186/s40249-022-00999-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 06/09/2022] [Indexed: 12/15/2022] Open
Abstract
Antibiotics have become a mainstay of healthcare in the past century due to their activity against pathogens. This manuscript reviews the impact of antibiotic use on the intestinal microbiota in the context of mass drug administration (MDA). The importance of the gut microbiota to human metabolism and physiology is now well established, and antibiotic exposure may impact host health via collateral effects on the microbiota and its functions. To gain further insight into how gut microbiota respond to antibiotic perturbation and the implications for public health, factors that influence the impact of antibiotic exposure on the microbiota, potential health outcomes of antibiotic-induced microbiota alterations, and strategies that have the potential to ameliorate these wider antibiotic-associated microbiota perturbations are also reviewed.
Collapse
Affiliation(s)
- Ethan K Gough
- Department of International Health, Human Nutrition Program, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
21
|
Cannon JL, Seabolt MH, Xu R, Montmayeur A, Suh SH, Diez-Valcarce M, Bucardo F, Becker-Dreps S, Vinjé J. Gut Microbiome Changes Occurring with Norovirus Infection and Recovery in Infants Enrolled in a Longitudinal Birth Cohort in Leon, Nicaragua. Viruses 2022; 14:v14071395. [PMID: 35891376 PMCID: PMC9323674 DOI: 10.3390/v14071395] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/07/2022] [Accepted: 06/13/2022] [Indexed: 02/05/2023] Open
Abstract
Noroviruses are associated with one fifth of diarrheal illnesses globally and are not yet preventable with vaccines. Little is known about the effects of norovirus infection on infant gut microbiome health, which has a demonstrated role in protecting hosts from pathogens and a possible role in oral vaccine performance. In this study, we characterized infant gut microbiome changes occurring with norovirus-associated acute gastroenteritis (AGE) and the extent of recovery. Metagenomic sequencing was performed on the stools of five infants participating in a longitudinal birth cohort study conducted in León, Nicaragua. Taxonomic and functional diversities of gut microbiomes were profiled at time points before, during, and after norovirus infection. Initially, the gut microbiomes resembled those of breastfeeding infants, rich in probiotic species. When disturbed by AGE, Gammaproteobacteria dominated, particularly Pseudomonas species. Alpha diversity increased but the genes involved in carbohydrate metabolism and glycan biosynthesis decreased. After the symptoms subsided, the gut microbiomes rebounded with their taxonomic and functional communities resembling those of the pre-infection microbiomes. In this study, during disruptive norovirus-associated AGE, the gut microbiome was temporarily altered, returning to a pre-infection composition a median of 58 days later. Our study provides new insights for developing probiotic treatments and furthering our understanding of the role that episodes of AGE have in shaping the infant gut microbiome, their long-term outcomes, and implications for oral vaccine effectiveness.
Collapse
Affiliation(s)
- Jennifer L. Cannon
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (R.X.); (A.M.); (S.H.S.); (M.D.-V.); (J.V.)
- CDC Foundation, Atlanta, GA 30329, USA
- Correspondence: ; Tel.: +1-404-639-2396
| | - Matthew H. Seabolt
- Office of Advanced Molecular Detection, National Center for Emerging & Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA;
- Leidos Inc., Reston, VA 20190, USA
| | - Ruijie Xu
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (R.X.); (A.M.); (S.H.S.); (M.D.-V.); (J.V.)
| | - Anna Montmayeur
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (R.X.); (A.M.); (S.H.S.); (M.D.-V.); (J.V.)
| | - Soo Hwan Suh
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (R.X.); (A.M.); (S.H.S.); (M.D.-V.); (J.V.)
- Ministry of Food and Drug Safety, Cheonju-Si 28159, Korea
| | - Marta Diez-Valcarce
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (R.X.); (A.M.); (S.H.S.); (M.D.-V.); (J.V.)
| | - Filemón Bucardo
- Center for Infectious Diseases Research, National Autonomous University of Nicaragua—León (UNAN-León), León 21000, Nicaragua;
| | - Sylvia Becker-Dreps
- Department of Family Medicine and Epidemiology, University of North Carolina Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Jan Vinjé
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (R.X.); (A.M.); (S.H.S.); (M.D.-V.); (J.V.)
| |
Collapse
|
22
|
Dey P, Ray Chaudhuri S. Cancer-Associated Microbiota: From Mechanisms of Disease Causation to Microbiota-Centric Anti-Cancer Approaches. BIOLOGY 2022; 11:757. [PMID: 35625485 PMCID: PMC9138768 DOI: 10.3390/biology11050757] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/08/2022] [Accepted: 05/12/2022] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori infection is the only well-established bacterial cause of cancer. However, due to the integral role of tissue-resident commensals in maintaining tissue-specific immunometabolic homeostasis, accumulated evidence suggests that an imbalance of tissue-resident microbiota that are otherwise considered as commensals, can also promote various types of cancers. Therefore, the present review discusses compelling evidence linking tissue-resident microbiota (especially gut bacteria) with cancer initiation and progression. Experimental evidence supporting the cancer-causing role of gut commensal through the modulation of host-specific processes (e.g., bile acid metabolism, hormonal effects) or by direct DNA damage and toxicity has been discussed. The opportunistic role of commensal through pathoadaptive mutation and overcoming colonization resistance is discussed, and how chronic inflammation triggered by microbiota could be an intermediate in cancer-causing infections has been discussed. Finally, we discuss microbiota-centric strategies, including fecal microbiota transplantation, proven to be beneficial in preventing and treating cancers. Collectively, this review provides a comprehensive understanding of the role of tissue-resident microbiota, their cancer-promoting potentials, and how beneficial bacteria can be used against cancers.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Saumya Ray Chaudhuri
- Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology, Chandigarh 160036, India;
| |
Collapse
|
23
|
The Concept of Intrauterine Programming and the Development of the Neonatal Microbiome in the Prevention of SARS-CoV-2 Infection. Nutrients 2022; 14:nu14091702. [PMID: 35565670 PMCID: PMC9104449 DOI: 10.3390/nu14091702] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/07/2022] [Accepted: 04/14/2022] [Indexed: 02/06/2023] Open
Abstract
The process of intrauterine programming is related to the quality of the microbiome formed in the fetus and the newborn. The implementation of probiotics, prebiotics, and psychobiotics shows immunomodulatory potential towards the organism, especially the microbiome of the pregnant woman and her child. Nutrigenomics, based on the observation of pregnant women and the developing fetus, makes it possible to estimate the biological effects of active dietary components on gene expression or silencing. Nutritional intervention for pregnant women should consider the nutritional status of the patient, biological markers, and the potential impact of dietary intervention on fetal physiology. The use of a holistic model of nutrition allows for appropriately targeted and effective dietary prophylaxis that can impact the physical and mental health of both the mother and the newborn. This model targets the regulation of the immune response of the pregnant woman and the newborn, considering the clinical state of the microbiota and the pathomechanism of the nervous system. Current scientific reports indicate the protective properties of immunobiotics (probiotics) about the reduction of the frequency of infections and the severity of the course of COVID-19 disease. The aim of this study was to test the hypothesis that intrauterine programming influences the development of the microbiome for the prevention of SARS-CoV-2 infection based on a review of research studies.
Collapse
|
24
|
Marcela K, de Meij GT, Fidelma F, Richard DJ, Mark WH, Ed KJ. How to: Clostridioides difficile infection in children. Clin Microbiol Infect 2022; 28:1085-1090. [DOI: 10.1016/j.cmi.2022.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 02/07/2022] [Accepted: 03/01/2022] [Indexed: 12/12/2022]
|
25
|
Lian S, Liu J, Wu Y, Xia P, Zhu G. Bacterial and Viral Co-Infection in the Intestine: Competition Scenario and Their Effect on Host Immunity. Int J Mol Sci 2022; 23:ijms23042311. [PMID: 35216425 PMCID: PMC8877981 DOI: 10.3390/ijms23042311] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/02/2022] [Accepted: 02/17/2022] [Indexed: 12/04/2022] Open
Abstract
Bacteria and viruses are both important pathogens causing intestinal infections, and studies on their pathogenic mechanisms tend to focus on one pathogen alone. However, bacterial and viral co-infections occur frequently in clinical settings, and infection by one pathogen can affect the severity of infection by another pathogen, either directly or indirectly. The presence of synergistic or antagonistic effects of two pathogens in co-infection can affect disease progression to varying degrees. The triad of bacterial–viral–gut interactions involves multiple aspects of inflammatory and immune signaling, neuroimmunity, nutritional immunity, and the gut microbiome. In this review, we discussed the different scenarios triggered by different orders of bacterial and viral infections in the gut and summarized the possible mechanisms of synergy or antagonism involved in their co-infection. We also explored the regulatory mechanisms of bacterial–viral co-infection at the host intestinal immune interface from multiple perspectives.
Collapse
Affiliation(s)
- Siqi Lian
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, Yangzhou 225009, China; (S.L.); (J.L.); (Y.W.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of China, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jiaqi Liu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, Yangzhou 225009, China; (S.L.); (J.L.); (Y.W.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of China, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yunping Wu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, Yangzhou 225009, China; (S.L.); (J.L.); (Y.W.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of China, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Pengpeng Xia
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, Yangzhou 225009, China; (S.L.); (J.L.); (Y.W.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of China, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence:
| | - Guoqiang Zhu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, Yangzhou 225009, China; (S.L.); (J.L.); (Y.W.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of China, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
26
|
Dynamic of the human gut microbiome under infectious diarrhea. Curr Opin Microbiol 2022; 66:79-85. [PMID: 35121284 DOI: 10.1016/j.mib.2022.01.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 11/20/2022]
Abstract
Despite the widespread implementation of sanitation, immunization and appropriate treatment, infectious diarrheal diseases still inflict a great health burden to children living in low resource settings. Conventional microbiology research in diarrhea have focused on the pathogen's biology and pathogenesis, but initial enteric infections could trigger subsequent perturbations in the gut microbiome, leading to short-term or long-term health effects. Conversely, such pre-existing perturbations could render children more vulnerable to enteropathogen colonization and diarrhea. Recent advances in DNA sequencing and bioinformatic analyses have been integrated in well-designed clinical and epidemiological studies, which allow us to track how the gut microbiome changes from disease onset to recovery. Here, we aim to summarize the current understanding on the diarrheal gut microbiome, stratified into different disease stages. Furthermore, we discuss how such perturbations could have impacts beyond an acute diarrhea episode, specifically on the child's nutritional status and the facilitation of antimicrobial resistance.
Collapse
|
27
|
George S, Aguilera X, Gallardo P, Farfán M, Lucero Y, Torres JP, Vidal R, O'Ryan M. Bacterial Gut Microbiota and Infections During Early Childhood. Front Microbiol 2022; 12:793050. [PMID: 35069488 PMCID: PMC8767011 DOI: 10.3389/fmicb.2021.793050] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota composition during the first years of life is variable, dynamic and influenced by both prenatal and postnatal factors, such as maternal antibiotics administered during labor, delivery mode, maternal diet, breastfeeding, and/or antibiotic consumption during infancy. Furthermore, the microbiota displays bidirectional interactions with infectious agents, either through direct microbiota-microorganism interactions or indirectly through various stimuli of the host immune system. Here we review these interactions during childhood until 5 years of life, focusing on bacterial microbiota, the most common gastrointestinal and respiratory infections and two well characterized gastrointestinal diseases related to dysbiosis (necrotizing enterocolitis and Clostridioides difficile infection). To date, most peer-reviewed studies on the bacterial microbiota in childhood have been cross-sectional and have reported patterns of gut dysbiosis during infections as compared to healthy controls; prospective studies suggest that most children progressively return to a "healthy microbiota status" following infection. Animal models and/or studies focusing on specific preventive and therapeutic interventions, such as probiotic administration and fecal transplantation, support the role of the bacterial gut microbiota in modulating both enteric and respiratory infections. A more in depth understanding of the mechanisms involved in the establishment and maintenance of the early bacterial microbiota, focusing on specific components of the microbiota-immunity-infectious agent axis is necessary in order to better define potential preventive or therapeutic tools against significant infections in children.
Collapse
Affiliation(s)
- Sergio George
- Host-Pathogen Interaction Laboratory, Microbiology and Mycology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Ximena Aguilera
- School of Medicine, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Pablo Gallardo
- Department of Pediatrics and Pediatric Surgery, Dr. Luis Calvo Mackenna Hospital, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Mauricio Farfán
- Department of Pediatrics and Pediatric Surgery, Dr. Luis Calvo Mackenna Hospital, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Yalda Lucero
- Host-Pathogen Interaction Laboratory, Microbiology and Mycology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile.,Department of Pediatrics and Pediatric Surgery, Dr. Roberto del Río Hospital, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juan Pablo Torres
- Host-Pathogen Interaction Laboratory, Microbiology and Mycology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile.,Department of Pediatrics and Pediatric Surgery, Dr. Luis Calvo Mackenna Hospital, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Roberto Vidal
- Microbiology and Mycology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile.,ANID - Millennium Science Initiative Program - Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
| | - Miguel O'Ryan
- Host-Pathogen Interaction Laboratory, Microbiology and Mycology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
28
|
Zhao W, Yu ML, Tao X, Cheng MH, Liu CC, Liu Y, Li YG. Analysis of the intestinal microbial community altered during rotavirus infection in suckling mice. Virol J 2021; 18:254. [PMID: 34930341 PMCID: PMC8686622 DOI: 10.1186/s12985-021-01727-5] [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: 07/08/2021] [Accepted: 12/10/2021] [Indexed: 11/10/2022] Open
Abstract
Background Rotavirus (RV) is a principal cause of diarrhea. However, there is a limited understanding regarding alteration of the gut microbial community structure and abundance during RV infection. This study was to characterize any potential associations between RV infection and the intestinal microbiota.
Methods Suckling mice were divided into normal group (NC) and infected group (RV) randomly. All of the suckling mice were euthanized four days post-RV infection. The virus titer was counted as fluorescent focus assay, and viral load was quantified by QPCR. Five sucking mice were randomly selected from each RV group and NC group for sample collection and pathological analysis. Mixed intestinal contents of the colon and rectum were collected from all of the suckling mice. To investigate the detailed relationship between RV infection and intestinal microbiota, the composition and distribution of intestinal microbiota from suckling mice were first analyzed using 16S rRNA sequencing technology. Results The results of the pathological characteristics showed that vacuolar degeneration, vasodilation, hyperemia, and destruction of the intestinal epithelium were apparent in the RV group. Representative genera from Lactobacillus and Fusobacterium were enriched in the NC group, while the Enterococcus and Escherichia/Shigella genera were enriched in the RV group. Helicobacter, Alloprevotrlla, Brevundimonas, Paenibacillus, and Parabacteroides were completely undetectable in the RV group. The predicted intestinal flora metabolic function results showed that “carbohydrate metabolism” and “lipid metabolism” pathways were significantly enriched within the NC group. A significant difference has been observed in the gut microbiota composition between the two groups. Conclusions Our results demonstrated a significant difference in the gut microbiota composition in RV-infected suckling mice as compared to the RV un-infected suckling mice group. This work may provide meaningful information regarding the bacterial genera changed during RV infection. Moreover, the changes in these bacteria may be related with the replication and pathogenesis of RV infection.
Collapse
Affiliation(s)
- Wei Zhao
- College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121200, Liaoning, People's Republic of China
| | - Mei Ling Yu
- College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121200, Liaoning, People's Republic of China
| | - XiaoLi Tao
- College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121200, Liaoning, People's Republic of China
| | - Mei Hui Cheng
- College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121200, Liaoning, People's Republic of China
| | - Chang Cheng Liu
- College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121200, Liaoning, People's Republic of China
| | - Yang Liu
- College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121200, Liaoning, People's Republic of China
| | - Yong Gang Li
- College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121200, Liaoning, People's Republic of China.
| |
Collapse
|
29
|
The effect of metformin on body mass index and metabolic parameters in non-diabetic HIV-positive patients: a meta-analysis. J Diabetes Metab Disord 2021; 20:1901-1911. [PMID: 34900832 DOI: 10.1007/s40200-021-00869-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/31/2021] [Indexed: 10/20/2022]
Abstract
We sought to evaluate the effetc of metformin on body mass index (BMI) and metabolic parameters in HIV-positive patients. We performed a comprehensive search through five major indexing databases, using keywords ("metformin" OR "dimethylguanylguanidine" OR "biguanide" OR "Glucophage") AND ("HIV" OR "human immunodeficiency virus" OR "AIDS" OR "Acquired immunodeficiency syndrome"), and all possible combinations until January 15, 2021. We measured standardized mean differences (SMD) and 95% confidence intervals (CI) for each outcome. We finally included 12 RCTs (577 participants, 274 in the metformin group and 303 in the comparators). Metformin did not significantly change BMI index compared to various comparators. Metformin generally improve LDL levels (SMD = 0.29, 95% CI: - 1.00 1.57, P = 0.01), HDL levels (SMD = - 0.15, 95% CI: - 0.72 0.41, P = 0.001), triglycerides values (SMD = 0.46, 95% CI: - 0.36 1.27, P < 0.00001), fasting glucose (SMD = - 0.82, 95% CI: - 1.80 0.15, P < 0.00001), insulin 120 min (SMD = - 0.82, 95% CI: - 1.59-0.04, P = 0.02), and glucose 120 min (SMD = - 1.24, 95% CI: - 2.57 0.10, P < 0.0001), but worsened total cholesterol values (SMD = 1.24, 95% CI: - 0.98 3.46, P = 0.0001). Metformin is safe for weight loss in obese people; however, this drug may not be suitable for everyone, especially those who are not overweight. Nevertheless the body of evidences may suggest that metformin had promising impacts on metabolic parameters in patients with both HIV, it is still unknown that such surrogate changes will translate to long-standing clinical advantages. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-021-00869-1.
Collapse
|
30
|
Chronic Intestinal Pseudo-Obstruction: Is There a Connection with Gut Microbiota? Microorganisms 2021; 9:microorganisms9122549. [PMID: 34946150 PMCID: PMC8703706 DOI: 10.3390/microorganisms9122549] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/23/2022] Open
Abstract
Chronic intestinal pseudo-obstruction (CIPO) is a rare clinical syndrome characterized by severe impairment of gastrointestinal (GI) motility, and its symptoms are suggestive of partial or complete intestinal obstruction in the absence of any lesion restricting the intestinal lumen. Diagnosis and therapy of CIPO patients still represent a significant challenge for clinicians, despite their efforts to improve diagnostic workup and treatment strategies for this disease. The purpose of this review is to better understand what is currently known about the relationship between CIPO patients and intestinal microbiota, with a focus on the role of the enteric nervous system (ENS) and the intestinal endocrine system (IES) in intestinal motility, underling the importance of further studies to deeply understand the causes of gut motility dysfunction in these patients.
Collapse
|
31
|
Lucero Y, Matson DO, Ashkenazi S, George S, O’Ryan M. Norovirus: Facts and Reflections from Past, Present, and Future. Viruses 2021; 13:v13122399. [PMID: 34960668 PMCID: PMC8707792 DOI: 10.3390/v13122399] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Human Norovirus is currently the main viral cause of acute gastroenteritis (AGEs) in most countries worldwide. Nearly 50 years after the discovery of the "Norwalk virus" by Kapikian and colleagues, the scientific and medical community continue to generate new knowledge on the full biological and disease spectrum of Norovirus infection. Nevertheless, several areas remain incompletely understood due to the serious constraints to effectively replicate and propagate the virus. Here, we present a narrated historic perspective and summarize our current knowledge, including insights and reflections on current points of interest for a broad medical community, including clinical and molecular epidemiology, viral-host-microbiota interactions, antivirals, and vaccine prototypes. We also include a reflection on the present and future impacts of the COVID-19 pandemic on Norovirus infection and disease.
Collapse
Affiliation(s)
- Yalda Lucero
- Microbiology and Mycology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (Y.L.); (S.G.)
- Hospital Dr. Roberto del Río Hospital, Department of Pediatrics and Pediatric Surgery (Northern Campus), Faculty of Medicine, Universidad de Chile, Santiago 8380418, Chile
- Clínica Alemana de Santiago, Faculty of Medicine, Universidad del Desarrollo-Clínica Alemana, Santiago 7650568, Chile
| | - David O. Matson
- Eastern Shore Health Department, Virginia Department of Public Health, Accomack County, VA 23301, USA;
| | - Shai Ashkenazi
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
- Department of Pediatrics A, Schneider Children’s Medical Center, Petach Tikva 49202, Israel
| | - Sergio George
- Microbiology and Mycology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (Y.L.); (S.G.)
| | - Miguel O’Ryan
- Microbiology and Mycology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (Y.L.); (S.G.)
- Correspondence:
| |
Collapse
|
32
|
Baradaran Ghavami S, Pourhamzeh M, Farmani M, Keshavarz H, Shahrokh S, Shpichka A, Asadzadeh Aghdaei H, Hakemi-Vala M, Hossein-khannazer N, Timashev P, Vosough M. Cross-talk between immune system and microbiota in COVID-19. Expert Rev Gastroenterol Hepatol 2021; 15:1281-1294. [PMID: 34654347 PMCID: PMC8567289 DOI: 10.1080/17474124.2021.1991311] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/06/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Human gut microbiota plays a crucial role in providing protective responses against pathogens, particularly by regulating immune system homeostasis. There is a reciprocal interaction between the gut and lung microbiota, called the gut-lung axis (GLA). Any alteration in the gut microbiota or their metabolites can cause immune dysregulation, which can impair the antiviral activity of the immune system against respiratory viruses such as severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2. AREAS COVERED This narrative review mainly outlines emerging data on the mechanisms underlying the interactions between the immune system and intestinal microbial dysbiosis, which is caused by an imbalance in the levels of essential metabolites. The authors will also discuss the role of probiotics in restoring the balance of the gut microbiota and modulation of cytokine storm. EXPERT OPINION Microbiota-derived signals regulate the immune system and protect different tissues during severe viral respiratory infections. The GLA's equilibration could help manage the mortality and morbidity rates associated with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Shaghayegh Baradaran Ghavami
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Mahsa Pourhamzeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Farmani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Hediye Keshavarz
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Shabnam Shahrokh
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Mojdeh Hakemi-Vala
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikoo Hossein-khannazer
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
33
|
Adeyemi OO, Alabi AS, Adeyemi OA, Talabi OT, Abidakun OM, Joel IY, Stonehouse NJ. Acute gastroenteritis and the usage pattern of antibiotics and traditional herbal medications for its management in a Nigerian community. PLoS One 2021; 16:e0257837. [PMID: 34607333 PMCID: PMC8490005 DOI: 10.1371/journal.pone.0257837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/12/2021] [Indexed: 11/23/2022] Open
Abstract
Acute gastroenteritis (AGE) is the highest cause of mortality worldwide in children under the age of 5 years, with the highest mortalities occurring in low-to-middle income countries. Treatment can involve use of unregulated herbal medication and antibiotics. A cross sectional study was carried out to investigate the use of antibiotics and traditional herbal medications in the management of AGE among Yòrùbá-speaking communities in Kwara State, Nigeria. Our findings suggest habitual use of antibiotics (54.6%) and herbal medication (42.5%) in the management of AGE with high levels of self-prescription of antibiotics (21.7%) and herbal medications (36.2%) within the community. Ethanolic extracts of selected herbal plants reported (i.e. Aristolochia ringens, Azadirachta indica, Chromolaena odorata, Etanda Africana, Ficus capensis, Ficus vogelii, Mangifera indica, Momordica charantia, Ocimum gratisimum, Senna alata, Sorghum bicolor and Vernonia amygdalina) were investigated for antibacterial properties, using bacteria known to be causative agents of AGE. Our findings showed that, with exception of Ficus vogelii, which enhanced bacterial growth, the plant extracts reported all showed some antibacterial activity. We further discuss our findings within a regulatory context, with the aim to guide the use of traditional and herbal medication in low-to medium income countries (LMICs) and reduce the potential risks associated with the development of antimicrobial resistance.
Collapse
Affiliation(s)
- Oluwapelumi Olufemi Adeyemi
- Department of Medical Microbiology and Parasitology, Faculty of Basic Clinical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, Garstang Building, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- * E-mail: (OOA); (NJS)
| | - Ade Stephen Alabi
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | | | - Olaoluwa Temitope Talabi
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | | | | | - Nicola J. Stonehouse
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, Garstang Building, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- * E-mail: (OOA); (NJS)
| |
Collapse
|
34
|
Significance of the Gut Microbiome for Viral Diarrheal and Extra-Intestinal Diseases. Viruses 2021; 13:v13081601. [PMID: 34452466 PMCID: PMC8402659 DOI: 10.3390/v13081601] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/29/2021] [Accepted: 08/09/2021] [Indexed: 12/13/2022] Open
Abstract
The composition of the mammalian gut microbiome is very important for the health and disease of the host. Significant correlations of particular gut microbiota with host immune responsiveness and various infectious and noninfectious host conditions, such as chronic enteric infections, type 2 diabetes, obesity, asthma, and neurological diseases, have been uncovered. Recently, research has moved on to exploring the causalities of such relationships. The metabolites of gut microbiota and those of the host are considered in a ‘holobiontic’ way. It turns out that the host’s diet is a major determinant of the composition of the gut microbiome and its metabolites. Animal models of bacterial and viral intestinal infections have been developed to explore the interrelationships of diet, gut microbiome, and health/disease phenotypes of the host. Dietary fibers can act as prebiotics, and certain bacterial species support the host’s wellbeing as probiotics. In cases of Clostridioides difficile-associated antibiotic-resistant chronic diarrhea, transplantation of fecal microbiomes has sometimes cured the disease. Future research will concentrate on the definition of microbial/host/diet interrelationships which will inform rationales for improving host conditions, in particular in relation to optimization of immune responses to childhood vaccines.
Collapse
|
35
|
Toro Monjaraz EM, Ignorosa Arellano KR, Loredo Mayer A, Palacios-González B, Cervantes Bustamante R, Ramírez Mayans JA. Gut Microbiota in Mexican Children With Acute Diarrhea: An Observational Study. Pediatr Infect Dis J 2021; 40:704-709. [PMID: 34250970 DOI: 10.1097/inf.0000000000003128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Acute diarrhea is the second leading cause of preventable mortality and morbidity in children worldwide. This study aimed to identify the main pathogens associated with acute diarrhea and to describe changes in gut microbiota in Mexican children. METHODS This single-center observational study included 30 children (6 months to 5 years old) with acute diarrhea who were referred to the Instituto Nacional de Pediatría of Mexico City and 15 healthy volunteers (control group). Stool samples at day 0 (D0) and day 15 (D15) were collected for identification of microorganisms (reverse transcriptase-polymerase chain reaction analyses with xTAG gastrointestinal pathogen panel multiplex assay) and microbiota analysis (16S gene amplification sequencing). Prescription decisions were made by the treating clinician. RESULTS The main pathogens identified were norovirus and Campylobacter jejuni (20% each). The majority of patients (n = 24) were prescribed Saccharomyces boulardii CNCM I-745 for treatment of acute diarrhea. Diarrheic episodes resolved within 1 week of treatment. Compared with D15 and control samples, D0 samples showed significantly lower alpha diversity and a clear shift in overall composition (beta diversity). Alpha diversity was significantly increased in S. boulardii-treated group between D0 and D15 to a level similar to that of control group. CONCLUSIONS In these children, acute diarrhea was accompanied by significant alterations in gut microbiota. S. boulardii CNCM I-745 treatment may facilitate gut microbiota restoration in children with acute diarrhea, mostly through improvements in alpha diversity.
Collapse
Affiliation(s)
| | | | - Alejandro Loredo Mayer
- From the Pediatric Gastroenterology and Nutrition Department, Instituto Nacional de Pediatría
| | - Berenice Palacios-González
- Scientific Bonding Unit, Medicine Faculty UNAM, Instituto Nacional De Medicina Genómica, Mexico City, Mexico
| | | | | |
Collapse
|
36
|
Indo Y, Kitahara S, Tomokiyo M, Araki S, Islam MA, Zhou B, Albarracin L, Miyazaki A, Ikeda-Ohtsubo W, Nochi T, Takenouchi T, Uenishi H, Aso H, Takahashi H, Kurata S, Villena J, Kitazawa H. Ligilactobacillus salivarius Strains Isolated From the Porcine Gut Modulate Innate Immune Responses in Epithelial Cells and Improve Protection Against Intestinal Viral-Bacterial Superinfection. Front Immunol 2021; 12:652923. [PMID: 34163470 PMCID: PMC8215365 DOI: 10.3389/fimmu.2021.652923] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/20/2021] [Indexed: 12/29/2022] Open
Abstract
Previously, we constructed a library of Ligilactobacillus salivarius strains from the intestine of wakame-fed pigs and reported a strain-dependent capacity to modulate IFN-β expression in porcine intestinal epithelial (PIE) cells. In this work, we further characterized the immunomodulatory activities of L. salivarius strains from wakame-fed pigs by evaluating their ability to modulate TLR3- and TLR4-mediated innate immune responses in PIE cells. Two strains with a remarkable immunomodulatory potential were selected: L. salivarius FFIG35 and FFIG58. Both strains improved IFN-β, IFN-λ and antiviral factors expression in PIE cells after TLR3 activation, which correlated with an enhanced resistance to rotavirus infection. Moreover, a model of enterotoxigenic E. coli (ETEC)/rotavirus superinfection in PIE cells was developed. Cells were more susceptible to rotavirus infection when the challenge occurred in conjunction with ETEC compared to the virus alone. However, L. salivarius FFIG35 and FFIG58 maintained their ability to enhance IFN-β, IFN-λ and antiviral factors expression in PIE cells, and to reduce rotavirus replication in the context of superinfection. We also demonstrated that FFIG35 and FFIG58 strains regulated the immune response of PIE cells to rotavirus challenge or ETEC/rotavirus superinfection through the modulation of negative regulators of the TLR signaling pathway. In vivo studies performed in mice models confirmed the ability of L. salivarius FFIG58 to beneficially modulate the innate immune response and protect against ETEC infection. The results of this work contribute to the understanding of beneficial lactobacilli interactions with epithelial cells and allow us to hypothesize that the FFIG35 or FFIG58 strains could be used for the development of highly efficient functional feed to improve immune health status and reduce the severity of intestinal infections and superinfections in weaned piglets.
Collapse
Affiliation(s)
- Yuhki Indo
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shugo Kitahara
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Mikado Tomokiyo
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shota Araki
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Md. Aminul Islam
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Department of Medicine, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Binghui Zhou
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Leonardo Albarracin
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Scientific Computing Laboratory, Computer Science Department, Faculty of Exact Sciences and Technology, National University of Tucuman, Tucuman, Argentina
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli, (CERELA-CONICET), Tucuman, Argentina
| | - Ayako Miyazaki
- Viral Diseases and Epidemiology Research Division, National Institute of Animal Health, NARO, Tsukuba, Japan
| | - Wakako Ikeda-Ohtsubo
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Tomonori Nochi
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Laboratory of Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Takato Takenouchi
- Animal Bioregulation Unit, Division of Animal Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Hirohide Uenishi
- Animal Bioregulation Unit, Division of Animal Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Hisashi Aso
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Laboratory of Animal Health Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Hideki Takahashi
- Laboratory of Plant Pathology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Plant Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shoichiro Kurata
- Laboratory of Molecular Genetics, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Julio Villena
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli, (CERELA-CONICET), Tucuman, Argentina
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
37
|
Julio-Pieper M, López-Aguilera A, Eyzaguirre-Velásquez J, Olavarría-Ramírez L, Ibacache-Quiroga C, Bravo JA, Cruz G. Gut Susceptibility to Viral Invasion: Contributing Roles of Diet, Microbiota and Enteric Nervous System to Mucosal Barrier Preservation. Int J Mol Sci 2021; 22:ijms22094734. [PMID: 33946994 PMCID: PMC8125429 DOI: 10.3390/ijms22094734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 02/08/2023] Open
Abstract
The gastrointestinal lumen is a rich source of eukaryotic and prokaryotic viruses which, together with bacteria, fungi and other microorganisms comprise the gut microbiota. Pathogenic viruses inhabiting this niche have the potential to induce local as well as systemic complications; among them, the viral ability to disrupt the mucosal barrier is one mechanism associated with the promotion of diarrhea and tissue invasion. This review gathers recent evidence showing the contributing effects of diet, gut microbiota and the enteric nervous system to either support or impair the mucosal barrier in the context of viral attack.
Collapse
Affiliation(s)
- Marcela Julio-Pieper
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340000, Chile; (A.L.-A.); (J.E.-V.); (J.A.B.)
- Correspondence:
| | - Alejandra López-Aguilera
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340000, Chile; (A.L.-A.); (J.E.-V.); (J.A.B.)
| | - Johana Eyzaguirre-Velásquez
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340000, Chile; (A.L.-A.); (J.E.-V.); (J.A.B.)
| | | | - Claudia Ibacache-Quiroga
- Centro de Micro-Bioinnovación (CMBi), Escuela de Nutrición y Dietética, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2340000, Chile;
| | - Javier A. Bravo
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340000, Chile; (A.L.-A.); (J.E.-V.); (J.A.B.)
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile;
| |
Collapse
|
38
|
Sohail MU, Al Khatib HA, Al Thani AA, Al Ansari K, Yassine HM, Al-Asmakh M. Microbiome profiling of rotavirus infected children suffering from acute gastroenteritis. Gut Pathog 2021; 13:21. [PMID: 33781328 PMCID: PMC8005861 DOI: 10.1186/s13099-021-00411-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
Background Rotavirus (RV) is a leading cause of pediatric diarrhea and mortality worldwide. The virus causes acute gastroenteritis characterized by moderate to severe vomiting, diarrhea, dehydration, and fever. Microbial dysbiosis caused by RV infection may significantly influence disease prognosis and the development of other chronic diseases. The gut microbiome plays a vital role in enteric immune response for rotavirus vaccine (RVV) that requires further elucidations. The current study evaluates the gut microbiome of RV positive children and compares gastroenteritis manifestation in children admitted to the Pediatric Emergency Centre, Hamad Medical Cooperation, Doha, Qatar. Stool samples were collected from thirty-nine RV positive and eight healthy control children. 16S rRNA sequence was performed using the Illumina MiSeq platform. Results The data demonstrated a significant increase in microbiome diversity denoted by higher relative abundances of phylum Proteobacteria (p = 0.031), Fusobacteria (p = 0.044) and genus Streptococcus (p ≤ 0.001) in the infected group relative to the control. Similarly, district clustering pattern (PERMANOVA p = 0.01) and higher species richness (Shannon entropy p = 0.018) were observed in the children who received two RVV doses compared with the non-vaccinated or single-dose groups. These microbiome changes were represented by over-abundance of phylum Bacteroidetes (p = 0.003) and Verrucomicrobia (p ≤ 0.001), and lower expression of family Enterobacteriaceae in two RVV doses group. However, microbiome composition was not associated with diarrhea, vomiting, and other parameters of gastroenteritis. Conclusions The observations assert significant microbial signatures of RVV, which is dose-dependent, and suggest manipulating these microbes as a novel approach for improving RVV efficacy. Further studies are warranted to investigate the immune status of these patients and mechanistic investigation to enhance RVV seroconversion.
Collapse
Affiliation(s)
- Muhammad U Sohail
- Proteomics Core, Weill Cornell Medicine-Qatar, P.O. Box 24811, Doha, Qatar
| | - Hebah A Al Khatib
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Asmaa A Al Thani
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar.,Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Khalid Al Ansari
- Emergency Medicine Department, Sidra Medicine, Qatar Foundation, Doha, Qatar
| | - Hadi M Yassine
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar. .,Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Maha Al-Asmakh
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar. .,Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar. .,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
39
|
Tiwari V, Anand Y, Gupta A, Divyaveer S, Bhargava V, Malik M, Gupta A, Bhalla AK, Rana DS. Etiological Spectrum of Infective Diarrhea in Renal Transplant Patient by Stool PCR: An Indian Perspective. Indian J Nephrol 2021; 31:245-253. [PMID: 34376938 PMCID: PMC8330656 DOI: 10.4103/ijn.ijn_169_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/19/2020] [Accepted: 07/05/2020] [Indexed: 11/04/2022] Open
Abstract
Introduction: Diarrhea is a common cause of morbidity and mortality among renal transplant patients. The etiological spectrum of pathogens varies with regional diversity, socioeconomic conditions, sanitation, and eating habits. We aimed to delineate the etiological profile of gastrointestinal pathogens in renal transplant patients using the stool Polymerase chain reaction. Methods: In this single-center, retrospective analysis of patients from January 2016 to January 2018, all renal transplant patients who were admitted with severe diarrhea and underwent the stool Polymerase chain reaction (PCR) were included. In the control group, we included patients from the general population who were admitted with similar complaints in the general medicine ward and underwent stool PCR over the same duration. Results: One hundred ten admissions occurred over 2 years in the transplant group. 86% of samples were positive for infection. More than one organism was seen in 68% of the patient. Norovirus was the most common organism isolated. Giardia lamblia with Norovirus was the most common coinfection among the transplant population. In the control group, 87% of samples tested positive, with 53% of patients having more than one organism. Enteroaggregative E. coli was the common organism, Enteroaggregative E. coli with Enteropathogenic E. coli and Enterotoxigenic E. coli were the most common organism in combination. Both the groups had similar incidence of infection with multiple organisms. Conclusion: The etiological profile of gastrointestinal pathogens differs significantly between the transplant and general population. Coinfections are common in both populations. Norovirus is the most common pathogen in the transplant population, presenting as isolated as well as in coinfections.
Collapse
Affiliation(s)
- Vaibhav Tiwari
- Department of Nephrology, Sir Ganga Ram Hospital, New Delhi, India
| | - Yogeshman Anand
- Department of Nephrology, Sir Ganga Ram Hospital, New Delhi, India
| | - Anurag Gupta
- Department of Nephrology, Sir Ganga Ram Hospital, New Delhi, India
| | | | - Vinant Bhargava
- Department of Nephrology, Sir Ganga Ram Hospital, New Delhi, India
| | - Manish Malik
- Department of Nephrology, Sir Ganga Ram Hospital, New Delhi, India
| | - Ashwani Gupta
- Department of Nephrology, Sir Ganga Ram Hospital, New Delhi, India
| | | | - D S Rana
- Department of Nephrology, Sir Ganga Ram Hospital, New Delhi, India
| |
Collapse
|
40
|
Sainz T, Delgado J, Mendez‐Echevarría A, Santiago B, Lopez‐Varela E, Aguilera‐Alonso D, Saavedra‐Lozano J, Rodríguez‐Fernández R, Holguín Á, Navarro ML, Muñoz‐Fernández MÁ, Rivero‐Calle I, Solana MJ, López‐Herce J, Calvo C. The clinical relevance of the microbiome when managing paediatric infectious diseases-Narrative review. Acta Paediatr 2021; 110:441-449. [PMID: 32961592 DOI: 10.1111/apa.15578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/18/2020] [Accepted: 09/15/2020] [Indexed: 11/28/2022]
Abstract
In recent years, the field of infectious diseases has been hit by the overwhelming amount of information generated while the human microbiome is being disentangled. Based on the interaction between the microbiota and the immune system, the implications regarding infectious diseases are probably major and remain a challenge. AIMS This review was conceived as a comprehensive tool to provide an overview of the available evidence regarding the influence of the microbiome on infectious diseases in children. METHODS We present the main findings aroused from microbiome research in prevention, diagnosis and treatment of infectious disease under a paediatric perspective, to inform clinicians of the potential relevance of microbiome-related knowledge for translation to clinical practice. RESULTS AND CONCLUSION The evidence shown in this review highlights the numerous research gaps ahead and supports the need to move forward to integrating the so-called microbiome thinking into our routine clinical practice.
Collapse
Affiliation(s)
- Talía Sainz
- Hospital La Paz and La Paz Research Intitute (IdiPAZ) Madrid Spain
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
| | - Jaime Delgado
- Hospital La Paz and La Paz Research Intitute (IdiPAZ) Madrid Spain
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
| | - Ana Mendez‐Echevarría
- Hospital La Paz and La Paz Research Intitute (IdiPAZ) Madrid Spain
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
| | - Begoña Santiago
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
- Hospital General Universitario Gregorio Marañón and Research Institute IISGM Madrid Spain
| | - Elisa Lopez‐Varela
- ISGlobal, Barcelona Centre for International Health ResearchHospital Clinic ‐ Universitat de Barcelona Barcelona Spain
| | - David Aguilera‐Alonso
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
- Hospital General Universitario Gregorio Marañón and Research Institute IISGM Madrid Spain
| | - Jesús Saavedra‐Lozano
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
- Hospital General Universitario Gregorio Marañón and Research Institute IISGM Madrid Spain
| | - Rosa Rodríguez‐Fernández
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
- Hospital General Universitario Gregorio Marañón and Research Institute IISGM Madrid Spain
| | - África Holguín
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
- Ramón y Cajal Research Institute (IRyCIS)‐CIBERESP in Hospital Ramón y Cajal Madrid Spain
| | - Marisa L. Navarro
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
- Hospital General Universitario Gregorio Marañón and Research Institute IISGM Madrid Spain
| | - María Ángeles Muñoz‐Fernández
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
- Hospital General Universitario Gregorio Marañón and Research Institute IISGM Madrid Spain
| | - Irene Rivero‐Calle
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
- Hospital Clínico Universitario de Santiago‐CHUS and Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - María José Solana
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
- Hospital General Universitario Gregorio Marañón and Research Institute IISGM Madrid Spain
| | - Jesús López‐Herce
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
- Hospital General Universitario Gregorio Marañón and Research Institute IISGM Madrid Spain
| | - Cristina Calvo
- Hospital La Paz and La Paz Research Intitute (IdiPAZ) Madrid Spain
- The Traslational Research Network of Pediatric Infectious Diseases (RITIP) Madrid Spain
| |
Collapse
|
41
|
Sadiq FA. Is it time for microbiome-based therapies in viral infections? Virus Res 2021; 291:198203. [PMID: 33132161 PMCID: PMC7580679 DOI: 10.1016/j.virusres.2020.198203] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 01/07/2023]
Abstract
Infectious diseases related to viruses, as well as bacterial pathogens, abound in all parts of the world, burdening health and economy. Thus, there is a dire need to find new prevention and treatment strategies to improve clinical practices related to viral infections. Human gut contains trillions of bacteria which have regulatory roles in immune development, homeostasis, and body metabolism. Today, it is difficult to find any prominent viral infection that hasn't had any link with the human gut microbiota. In this opinion-based review article, I argued the significance of manipulating human gut microbiota as novel therapeutics through probiotics or FMT in alleviating complexities related to viral infections, and pinpointed bottlenecks involved in this research.
Collapse
Affiliation(s)
- Faizan Ahmed Sadiq
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, 1800 Lihu Avenue, 214122, China.
| |
Collapse
|
42
|
Din AU, Mazhar M, Waseem M, Ahmad W, Bibi A, Hassan A, Ali N, Gang W, Qian G, Ullah R, Shah T, Ullah M, Khan I, Nisar MF, Wu J. SARS-CoV-2 microbiome dysbiosis linked disorders and possible probiotics role. Biomed Pharmacother 2021; 133:110947. [PMID: 33197765 PMCID: PMC7657099 DOI: 10.1016/j.biopha.2020.110947] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/21/2020] [Accepted: 10/25/2020] [Indexed: 01/07/2023] Open
Abstract
In December 2019, a pneumonia outbreak of unknown etiology was reported which caused panic in Wuhan city of central China, which was later identified as Coronavirus disease (COVID-19) caused by a novel coronavirus, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) by the Chinese Centre for Disease Control and Prevention (CDC) and WHO. To date, the SARS-CoV-2 spread has already become a global pandemic with a considerable death toll. The associated symptoms of the COVID-19 infection varied with increased inflammation as an everyday pathological basis. Among various other symptoms such as fever, cough, lethargy, gastrointestinal (GI) symptoms included diarrhea and IBD with colitis, have been reported. Currently, there is no sole cure for COVID-19, and researchers are actively engaged to search out appropriate treatment and develop a vaccine for its prevention. Antiviral for controlling viral load and corticosteroid therapy for reducing inflammation seems to be inadequate to control the fatality rate. Based on the available related literature, which documented GI symptoms with diarrhea, inflammatory bowel diseases (IBD) with colitis, and increased deaths in the intensive care unit (ICU), conclude that dysbiosis occurs during SARS-COV-2 infection as the gut-lung axis cannot be ignored. As probiotics play a therapeutic role for GI, IBD, colitis, and even in viral infection. So, we assume that the inclusion of studies to investigate gut microbiome and subsequent therapies such as probiotics might help decrease the inflammatory response of viral pathogenesis and respiratory symptoms by strengthening the host immune system, amelioration of gut microbiome, and improvement of gut barrier function.
Collapse
Affiliation(s)
- Ahmad Ud Din
- Drug Discovery Research Center, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Maryam Mazhar
- Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Muhammed Waseem
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Horticulture, South China Agricultural University, Guangzhou, 510642, China
| | - Waqar Ahmad
- Drug Discovery Research Center, Southwest Medical University, Luzhou, 646000, Sichuan, China; College of Marine Life Sciences and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Asma Bibi
- Institute of Zoonosis Anhui Medical University, Hefei Anhui, 230032, China
| | - Adil Hassan
- Key Laboratory for Bio-rheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Niaz Ali
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bio-resources, College of Life Science and Technology, Guangxi University, 100 Daxue Road, Nanning, 530004, Guangxi, China
| | - Wang Gang
- Drug Discovery Research Center, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Gao Qian
- Drug Discovery Research Center, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Razi Ullah
- Key Laboratory for Bio-rheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Tariq Shah
- State Key Laboratory of Grassland Agro-Ecosystem, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Mehraj Ullah
- Department of Biotechnology School of Fermentation Engineering Tianjin University of Science and Technology China, China
| | - Israr Khan
- School of Life Sciences, Lanzhou University, China
| | - Muhammad Farrukh Nisar
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, 63100, Pakistan
| | - Jianbo Wu
- Drug Discovery Research Center, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
43
|
Olshan KL, Leonard MM, Serena G, Zomorrodi AR, Fasano A. Gut microbiota in Celiac Disease: microbes, metabolites, pathways and therapeutics. Expert Rev Clin Immunol 2020; 16:1075-1092. [PMID: 33103934 DOI: 10.1080/1744666x.2021.1840354] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Current evidence supports a vital role of the microbiota on health outcomes, with alterations in an otherwise healthy balance linked to chronic medical conditions like celiac disease (CD). Recent advances in microbiome analysis allow for unparalleled profiling of the microbes and metabolites. With the growing volume of data available, trends are emerging that support a role for the gut microbiota in CD pathogenesis. AREAS COVERED In this article, the authors review the relationship between factors such as genes and antibiotic exposure on CD onset and the intestinal microbiota. The authors also review other microbiota within the human body (like the oropharynx) that may play a role in CD pathogenesis. Finally, the authors discuss implications for disease modification and the ultimate goal of prevention. The authors reviewed literature from PubMed, EMBASE, and Web of Science. EXPERT OPINION CD serves as a unique opportunity to explore the role of the intestinal microbiota on the development of chronic autoimmune disease. While research to date provides a solid foundation, most studies have been case-control and thus do not have capacity to explore the mechanistic role of the microbiota in CD onset. Further longitudinal studies and integrated multi-omics are necessary for investigating CD pathogenesis.
Collapse
Affiliation(s)
- Katherine L Olshan
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Gloria Serena
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Ali R Zomorrodi
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,European Biomedical Research Institute of Salerno (EBRIS) , Salerno, Italy
| |
Collapse
|
44
|
Walters WA, Reyes F, Soto GM, Reynolds ND, Fraser JA, Aviles R, Tribble DR, Irvin AP, Kelley-Loughnane N, Gutierrez RL, Riddle MS, Ley RE, Goodson MS, Simons MP. Epidemiology and associated microbiota changes in deployed military personnel at high risk of traveler's diarrhea. PLoS One 2020; 15:e0236703. [PMID: 32785284 PMCID: PMC7423091 DOI: 10.1371/journal.pone.0236703] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Travelers’ diarrhea (TD) is the most prevalent illness encountered by deployed military personnel and has a major impact on military operations, from reduced job performance to lost duty days. Frequently, the etiology of TD is unknown and, with underreporting of cases, it is difficult to accurately assess its impact. An increasing number of ailments include an altered or aberrant gut microbiome. To better understand the relationships between long-term deployments and TD, we studied military personnel during two nine-month deployment cycles in 2015–2016 to Honduras. To collect data on the prevalence of diarrhea and impact on duty, a total of 1173 personnel completed questionnaires at the end of their deployment. 56.7% reported reduced performance and 21.1% reported lost duty days. We conducted a passive surveillance study of all cases of diarrhea reporting to the medical unit with 152 total cases and a similar pattern of etiology. Enteroaggregative E. coli (EAEC, 52/152), enterotoxigenic E. coli (ETEC, 50/152), and enteropathogenic E. coli (EPEC, 35/152) were the most prevalent pathogens detected. An active longitudinal surveillance of 67 subjects also identified diarrheagenic E. coli as the primary etiology (7/16 EPEC, 7/16 EAEC, and 6/16 ETEC). Eleven subjects were recruited into a nested longitudinal substudy to examine gut microbiome changes associated with deployment. A 16S rRNA amplicon survey of fecal samples showed differentially abundant baseline taxa for subjects who contracted TD versus those who did not, as well as detection of taxa positively associated with self-reported gastrointestinal distress. Disrupted microbiota was also qualitatively observable for weeks preceding and following the incidents of TD. These findings illustrate the complex etiology of diarrhea amongst military personnel in deployed settings and its impacts on job performance. Potential factors of resistance or susceptibility can provide a foundation for future clinical trials to evaluate prevention and treatment strategies.
Collapse
Affiliation(s)
| | | | - Giselle M. Soto
- U.S. Naval Medical Research Unit No. 6 (NAMRU-6),Callao, Lima, Peru
| | - Nathanael D. Reynolds
- Infectious Diseases Directorate, U.S. Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Jamie A. Fraser
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States of America
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States of America
| | | | - David R. Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States of America
| | - Adam P. Irvin
- 711 Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH, United States of America
| | - Nancy Kelley-Loughnane
- 711 Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH, United States of America
| | - Ramiro L. Gutierrez
- Infectious Diseases Directorate, U.S. Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Mark S. Riddle
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States of America
| | - Ruth E. Ley
- Max Planck Institute for Developmental Biology, Tuebingen, Germany
| | - Michael S. Goodson
- 711 Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH, United States of America
- * E-mail:
| | - Mark P. Simons
- Infectious Diseases Directorate, U.S. Naval Medical Research Center, Silver Spring, MD, United States of America
| |
Collapse
|
45
|
Eetemadi A, Rai N, Pereira BMP, Kim M, Schmitz H, Tagkopoulos I. The Computational Diet: A Review of Computational Methods Across Diet, Microbiome, and Health. Front Microbiol 2020; 11:393. [PMID: 32318028 PMCID: PMC7146706 DOI: 10.3389/fmicb.2020.00393] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Food and human health are inextricably linked. As such, revolutionary impacts on health have been derived from advances in the production and distribution of food relating to food safety and fortification with micronutrients. During the past two decades, it has become apparent that the human microbiome has the potential to modulate health, including in ways that may be related to diet and the composition of specific foods. Despite the excitement and potential surrounding this area, the complexity of the gut microbiome, the chemical composition of food, and their interplay in situ remains a daunting task to fully understand. However, recent advances in high-throughput sequencing, metabolomics profiling, compositional analysis of food, and the emergence of electronic health records provide new sources of data that can contribute to addressing this challenge. Computational science will play an essential role in this effort as it will provide the foundation to integrate these data layers and derive insights capable of revealing and understanding the complex interactions between diet, gut microbiome, and health. Here, we review the current knowledge on diet-health-gut microbiota, relevant data sources, bioinformatics tools, machine learning capabilities, as well as the intellectual property and legislative regulatory landscape. We provide guidance on employing machine learning and data analytics, identify gaps in current methods, and describe new scenarios to be unlocked in the next few years in the context of current knowledge.
Collapse
Affiliation(s)
- Ameen Eetemadi
- Department of Computer Science, University of California, Davis, Davis, CA, United States
- Genome Center, University of California, Davis, Davis, CA, United States
| | - Navneet Rai
- Genome Center, University of California, Davis, Davis, CA, United States
| | - Beatriz Merchel Piovesan Pereira
- Genome Center, University of California, Davis, Davis, CA, United States
- Department of Microbiology, University of California, Davis, Davis, CA, United States
| | - Minseung Kim
- Department of Computer Science, University of California, Davis, Davis, CA, United States
- Genome Center, University of California, Davis, Davis, CA, United States
- Process Integration and Predictive Analytics (PIPA LLC), Davis, CA, United States
| | - Harold Schmitz
- Graduate School of Management, University of California, Davis, Davis, CA, United States
| | - Ilias Tagkopoulos
- Department of Computer Science, University of California, Davis, Davis, CA, United States
- Genome Center, University of California, Davis, Davis, CA, United States
- Process Integration and Predictive Analytics (PIPA LLC), Davis, CA, United States
| |
Collapse
|
46
|
Woodward SE, Krekhno Z, Finlay BB. Here, there, and everywhere: How pathogenicEscherichia colisense and respond to gastrointestinal biogeography. Cell Microbiol 2019; 21:e13107. [DOI: 10.1111/cmi.13107] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Sarah E. Woodward
- Department of Microbiology and ImmunologyUniversity of British Columbia Vancouver British Columbia Canada
- Michael Smith LaboratoriesUniversity of British Columbia Vancouver British Columbia Canada
| | - Zakhar Krekhno
- Department of Microbiology and ImmunologyUniversity of British Columbia Vancouver British Columbia Canada
- Michael Smith LaboratoriesUniversity of British Columbia Vancouver British Columbia Canada
| | - B. Brett Finlay
- Department of Microbiology and ImmunologyUniversity of British Columbia Vancouver British Columbia Canada
- Michael Smith LaboratoriesUniversity of British Columbia Vancouver British Columbia Canada
- Department of Biochemistry and Molecular BiologyUniversity of British Columbia Vancouver British Columbia Canada
| |
Collapse
|
47
|
Adenovirus infection is associated with altered gut microbial communities in a non-human primate. Sci Rep 2019; 9:13410. [PMID: 31527752 PMCID: PMC6746978 DOI: 10.1038/s41598-019-49829-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
Adenovirus (AdV) infections are one of the main causes of diarrhea in young children. Enteric AdVs probably disrupt gut microbial defences, which can result in diarrhea. To understand the role of the gut microbiome in AdV-induced pathologies, we investigated the gut microbiome of a naturally AdV-infected non-human primate species, the Malagasy mouse lemur (Microcebus griseorufus), which represents an important model in understanding the evolution of diseases. We observed that AdV infection is associated with disruption of the gut microbial community composition. In AdV+ lemurs, several commensal taxa essential for a healthy gut microbiome decreased, whereas genera containing potential pathogens, such as Neisseria, increased in abundance. Microbial co-occurrence networks revealed a loss of important microbial community interactions in AdV+ lemurs and an overrepresentation of Prevotellaceae. The observation of enteric virus-associated loss of commensal bacteria and associated shifts towards pathobionts may represent the missing link for a better understanding of AdV-induced effects in humans, and also for their potential as drivers of co-infections, an area of research that has been largely neglected so far.
Collapse
|
48
|
Abstract
Antibiotics during infancy, delivery, and breastfeeding affect the intestinal microbiota in early life and is associated with allergic disease. Gastroenteritis (GE) during infancy also affects intestinal microbiota in early life, however, its relationship to allergic disease has not been investigated.Data of 45,499 males and 49,430 females, from birth to 5 years of age, were collected from a national database in Taiwan. Subjects were categorized into early GE (GE within 0-6 months) and non-early GE group (no GE within 0-6 months). The rates of asthma (AS), allergic rhinitis (AR), and atopic dermatitis (AD) over 5 years were evaluated and compared between the groups. In patients with AS, AR, and AD, the number of clinical visits and drug prescriptions for the allergic disease was also evaluated to assess the effect of early GE on allergic disease.After adjusting for the effect of GE in later life and other factors, the rates of AS [OR (odds ratio) 1.54, 95% confidence interval (CI) 1.48-1.60], AR [OR 1.49, 95% CI 1.45-1.54], and AD [OR 1.40, 95% CI 1.33-1.47] were higher in the early GE group than in the non-early GE group. The magnitude of the increase was higher in females than in males. In those with AS, AR, and AD, the number of clinical visits and drug prescriptions was not different between the early GE and non-early GE groups. In children with early GE, good control of GE in the following years lowered the rate of allergic disease.Early-life GE was associated with increased rates of AS, AR, and AD in later life and this was trend more prominent in females.
Collapse
Affiliation(s)
- Hui-Hsien Pan
- School of Medicine, Chung Shan Medical University
- Division of Allergy, Asthma and Rheumatology, Department of pediatrics, Chung Shan Medical University Hospital
| | - Ko-Huang Lue
- School of Medicine, Chung Shan Medical University
- University President, Chung Shan Medical University, Taichung, Taiwan
| | - Hai-Lun Sun
- School of Medicine, Chung Shan Medical University
- Division of Allergy, Asthma and Rheumatology, Department of pediatrics, Chung Shan Medical University Hospital
| | - Min-Sho Ku
- School of Medicine, Chung Shan Medical University
- Division of Allergy, Asthma and Rheumatology, Department of pediatrics, Chung Shan Medical University Hospital
| |
Collapse
|