451
|
Chorna N, Godoy-Vitorino F. A Protocol for the Multi-Omic Integration of Cervical Microbiota and Urine Metabolomics to Understand Human Papillomavirus (HPV)-Driven Dysbiosis. Biomedicines 2020; 8:biomedicines8040081. [PMID: 32276347 PMCID: PMC7235793 DOI: 10.3390/biomedicines8040081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/20/2022] Open
Abstract
The multi-omic integration of microbiota data with metabolomics has gained popularity. This protocol is based on a human multi-omics study, integrating cervicovaginal microbiota, HPV status and neoplasia, with urinary metabolites. Indeed, to understand the biology of the infections and to develop adequate interventions for cervical cancer prevention, studies are needed to characterize in detail the cervical microbiota and understand the systemic metabolome. This article is a detailed protocol for the multi-omic integration of cervical microbiota and urine metabolome to shed light on the systemic effects of cervical dysbioses associated with Human Papillomavirus (HPV) infections. This methods article suggests detailed sample collection and laboratory processes of metabolomics, DNA extraction for microbiota, HPV typing, and the bioinformatic analyses of the data, both to characterize the metabolome, the microbiota, and joint multi-omic analyses, useful for the development of new point-of-care diagnostic tests based on these approaches.
Collapse
Affiliation(s)
- Nataliya Chorna
- Department of Biochemistry, UPR School of Medicine, 00921 San Juan, Puerto Rico;
| | - Filipa Godoy-Vitorino
- Department of Microbiology & Medical Zoology, UPR School of Medicine, 00921 San Juan, Puerto Rico
- Correspondence: ; Tel.: +1-787-758-2525 (ext. 2096)
| |
Collapse
|
452
|
Hughes KR, Schofield Z, Dalby MJ, Caim S, Chalklen L, Bernuzzi F, Alcon-Giner C, Le Gall G, Watson AJM, Hall LJ. The early life microbiota protects neonatal mice from pathological small intestinal epithelial cell shedding. FASEB J 2020; 34:7075-7088. [PMID: 32253791 PMCID: PMC7610993 DOI: 10.1096/fj.202000042r] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 12/16/2022]
Abstract
The early life gut microbiota plays a crucial role in regulating and maintaining the intestinal barrier, with disturbances in these communities linked to dysregulated renewal and replenishment of intestinal epithelial cells. Here we sought to determine pathological cell shedding outcomes throughout the postnatal developmental period, and which host and microbial factors mediate these responses. Surprisingly, neonatal mice (Day 14 and 21) were highly refractory to induction of cell shedding after intraperitoneal administration of liposaccharide (LPS), with Day 29 mice showing strong pathological responses, more similar to those observed in adult mice. These differential responses were not linked to defects in the cellular mechanisms and pathways known to regulate cell shedding responses. When we profiled microbiota and metabolites, we observed significant alterations. Neonatal mice had high relative abundances of Streptococcus, Escherichia, and Enterococcus and increased primary bile acids. In contrast, older mice were dominated by Candidatus Arthromitus, Alistipes, and Lachnoclostridium, and had increased concentrations of SCFAs and methyamines. Antibiotic treatment of neonates restored LPS-induced small intestinal cell shedding, whereas adult fecal microbiota transplant alone had no effect. Our findings further support the importance of the early life window for microbiota-epithelial interactions in the presence of inflammatory stimuli and highlights areas for further investigation.
Collapse
Affiliation(s)
- Kevin R Hughes
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich, UK.,Norwich Medical School, University of East Anglia, Norwich, UK
| | - Zoe Schofield
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich, UK
| | - Matthew J Dalby
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich, UK
| | - Shabhonam Caim
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich, UK
| | - Lisa Chalklen
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich, UK
| | | | | | - Gwénaëlle Le Gall
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich, UK.,Norwich Medical School, University of East Anglia, Norwich, UK
| | | | - Lindsay J Hall
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich, UK.,Norwich Medical School, University of East Anglia, Norwich, UK
| |
Collapse
|
453
|
Derilus D, Godoy-Vitorino F, Rosado H, Agosto E, Dominguez-Bello MG, Cavallin H. An in-depth survey of the microbial landscape of the walls of a neonatal operating room. PLoS One 2020; 15:e0230957. [PMID: 32243474 PMCID: PMC7122808 DOI: 10.1371/journal.pone.0230957] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 03/12/2020] [Indexed: 12/22/2022] Open
Abstract
Bacteria found in operating rooms (ORs) might be clinically relevant since they could pose a threat to patients. In addition, C-sections operations are performed in ORs that provide the first environment and bacterial exposure to the sterile newborns that are extracted directly from the uterus to the OR air. Considering that at least one third of neonates in the US are born via C-section delivery (and more than 50% of all deliveries in some countries), understanding the distribution of bacterial diversity in ORs is critical to better understanding the contribution of the OR microbiota to C-section- associated inflammatory diseases. Here, we mapped the bacteria contained in an OR after a procedure was performed; we sampled grids of 60x60 cm across walls and wall-adjacent floors and sequenced the V4 region of 16S rRNA gene from 260 samples. The results indicate that bacterial communities changed significantly (ANOSIM, p-value < 0.001) with wall height, with an associated reduction of alpha diversity (t-test, p-value <0.05). OR walls contained high proportions of Proteobacteria, Firmicutes, and Actinobacteria, with Proteobacteria and Bacteroidetes being the highest in floors and lowest in the highest wall sites. Members of Firmicutes, Deinococcus-thermus, and Actinobacteria increased with wall height. Source-track analysis estimate that human skin is the major source contributing to bacterial composition in the OR walls, with an increase of bacteria related to human feces in the lowest walls and airborne bacteria in the highest wall sites. The results show that bacterial exposure in ORs varies spatially, and evidence exposure of C-section born neonates to human bacteria that remain on the floors and walls, possibly accumulated from patients, health, and cleaning staff.
Collapse
Affiliation(s)
- Dieunel Derilus
- Department of Environmental Sciences, University of Puerto Rico, San Juan, Puerto Rico, United States of America
| | - Filipa Godoy-Vitorino
- Department of Microbiology and Medical Zoology, University of Puerto-Rico-School of Medicine, San Juan, Puerto Rico, United States of America
| | - Hebe Rosado
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Edgardo Agosto
- School of Architecture, University of Puerto Rico, San Juan, Puerto Rico, United States of America
| | - Maria Gloria Dominguez-Bello
- Department of Biochemistry and Microbiology, and of Anthropology, and the New Jersey Institute for Food Nutrition and Health, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Humberto Cavallin
- School of Architecture, University of Puerto Rico, San Juan, Puerto Rico, United States of America
| |
Collapse
|
454
|
Skonieczna-Żydecka K, Janda K, Kaczmarczyk M, Marlicz W, Łoniewski I, Łoniewska B. The Effect of Probiotics on Symptoms, Gut Microbiota and Inflammatory Markers in Infantile Colic: A Systematic Review, Meta-Analysis and Meta-Regression of Randomized Controlled Trials. J Clin Med 2020; 9:E999. [PMID: 32252419 PMCID: PMC7231167 DOI: 10.3390/jcm9040999] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/25/2020] [Accepted: 03/30/2020] [Indexed: 12/24/2022] Open
Abstract
Immaturity in digestive-tract motor function and altered intestinal microbiome may play roles in pathogenesis of infantile colic. We assessed the impact of probiotic therapy on crying duration day, in newborns experiencing colic attacks. The PubMed, Embase, Cinnahl, Web of Science databases, and a clinical trials registry (ClinicalTrials.gov) were searched from inception until 12/02/2020. Random-effects meta-analyses were used to derive standardized mean differences/differences in means and risk ratios. We included 16 studies, which involved 1319 newborns aged up to 6 months. Lactobacillus reuteri strain DSM17938 was administered predominantly (n = 10). Probiotic intervention reduced the duration of crying (standardized mean difference = -2.012, 95% confidence interval: -2.763 to -1.261, z = -5.25, p < 0.0001). The probability of at least a 50% reduction in crying duration was at least 1.98 times higher in the intervention group than in controls (Z = 4.80, p < 0.0001). The effects of the intervention were not significantly affected by the risk of bias assessment, percentage of breastfed infants, and duration of the study. In 11 studies, data concerning gut microbiota composition and function and/or immunological markers were given. Probiotics significantly shortened the crying duration, but a causal relationship between the modulatory effect of probiotics on microbiota and the immune system has not been confirmed.
Collapse
Affiliation(s)
- Karolina Skonieczna-Żydecka
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (K.S.-Ż.); (K.J.); (I.Ł.)
| | - Katarzyna Janda
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (K.S.-Ż.); (K.J.); (I.Ł.)
| | - Mariusz Kaczmarczyk
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland;
| | - Wojciech Marlicz
- Department of Gastroenterology, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland;
| | - Igor Łoniewski
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (K.S.-Ż.); (K.J.); (I.Ł.)
| | - Beata Łoniewska
- Department of Neonatal Diseases, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| |
Collapse
|
455
|
El Bairi K, Jabi R, Trapani D, Boutallaka H, Ouled Amar Bencheikh B, Bouziane M, Amrani M, Afqir S, Maleb A. Can the microbiota predict response to systemic cancer therapy, surgical outcomes, and survival? The answer is in the gut. Expert Rev Clin Pharmacol 2020; 13:403-421. [PMID: 32308061 DOI: 10.1080/17512433.2020.1758063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The gut microbiota seems to play a key role in tumorigenesis, across various hallmarks of cancer. Recent evidence suggests its potential use as a biomarker predicting drug response and adding prognostic information, generally in the context of immuno-oncology. AREAS COVERED In this review, we focus on the modulating effects of gut microbiota dysbiosis on various anticancer molecules used in practice, including cytotoxic and immune-modulating agents, primarily immune-checkpoint inhibitors (ICI). Pubmed/Medline-based literature search was conducted to find potential original studies that discuss gut microbiota as a prognostic and predictive biomarker for cancer therapy. We also looked at the US ClinicalTrials.gov website to find additional studies particularly ongoing human clinical trials. EXPERT COMMENTARY Sequencing of stool-derived materials and tissue samples from cancer patients and animal models has shown a significant enrichment of various bacteria such as Fusobacterium nucleatum and Bacteroides fragilis were associated with resistant disease and poorer outcomes. Gut microbiota was also found to be associated with surgical outcomes and seems to play a significant role in anastomotic leak (ATL) after surgery mainly by collagen breakdown. However, this research field is just at the beginning and the current findings are not yet ready to change clinical practice.
Collapse
Affiliation(s)
- Khalid El Bairi
- Cancer Biomarkers Working Group, Mohamed Ist University , Oujda, Morocco
- Faculty of Medicine and Pharmacy, Mohamed Ist University , Oujda, Morocco
| | - Rachid Jabi
- Faculty of Medicine and Pharmacy, Mohamed Ist University , Oujda, Morocco
- Department of Visceral Surgery, Mohamed VI University Hospital , Oujda, Morocco
| | - Dario Trapani
- Department of Haematology and Oncology, European Institute of Oncology, IEO, IRCCS, University of Milano , Milan, Italy
| | - Hanae Boutallaka
- Department of Gastroenterology and Digestive Endoscopy, Mohamed V Military Teaching Hospital of Rabat, Mohamed V University , Rabat, Morocco
| | | | - Mohammed Bouziane
- Faculty of Medicine and Pharmacy, Mohamed Ist University , Oujda, Morocco
- Department of Visceral Surgery, Mohamed VI University Hospital , Oujda, Morocco
| | - Mariam Amrani
- Department of Pathology, National Institute of Oncology, Faculty of Medicine and Pharmacy, Mohamed V University , Rabat, Morocco
| | - Said Afqir
- Cancer Biomarkers Working Group, Mohamed Ist University , Oujda, Morocco
- Faculty of Medicine and Pharmacy, Mohamed Ist University , Oujda, Morocco
- Department of Medical Oncology, Mohamed VI University Hospital , Oujda, Morocco
| | - Adil Maleb
- Faculty of Medicine and Pharmacy, Mohamed Ist University , Oujda, Morocco
- Department of Microbiology, Mohamed VI University Hospital , Oujda, Morocco
| |
Collapse
|
456
|
Boem F, Nannini G, Amedei A. Not just 'immunity': how the microbiota can reshape our approach to cancer immunotherapy. Immunotherapy 2020; 12:407-416. [PMID: 32266849 DOI: 10.2217/imt-2019-0192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/23/2020] [Indexed: 11/21/2022] Open
Abstract
Cancer immunotherapy refers to a set of approaches aiming at enhancing the immune system to fight cancer growth and spread. This variety of therapeutic approaches, especially those inhibiting immune checkpoints, have shown very promising results. Nevertheless, patients may respond differently to treatments and the efficacy of immunotherapy seems to be dependent on several factors that go beyond the molecular targeting of immune cells modulation. Here, we review how the activity of gut microbiota appears to be crucial in determining the effectiveness of some immunotherapeutic treatments, fostering or impeding the conditions under which treatments can work or not. Moreover, we discuss how these findings suggest not only extending the range of immunotherapeutic approaches but also reshaping our understanding of immunotherapy itself.
Collapse
Affiliation(s)
- Federico Boem
- Department of Oncology and Hemato-Oncology, University of Milan, Milan 20122, Italy
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
- SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Florence 50134, Italy
| |
Collapse
|
457
|
Philips CA, Augustine P, Yerol PK, Ramesh GN, Ahamed R, Rajesh S, George T, Kumbar S. Modulating the Intestinal Microbiota: Therapeutic Opportunities in Liver Disease. J Clin Transl Hepatol 2020; 8:87-99. [PMID: 32274349 PMCID: PMC7132020 DOI: 10.14218/jcth.2019.00035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/11/2019] [Accepted: 10/27/2019] [Indexed: 12/11/2022] Open
Abstract
Gut microbiota has been demonstrated to have a significant impact on the initiation, progression and development of complications associated with multiple liver diseases. Notably, nonalcoholic fatty liver diseases, including nonalcoholic steatohepatitis and cirrhosis, severe alcoholic hepatitis, primary sclerosing cholangitis and hepatic encephalopathy, have strong links to dysbiosis - or a pathobiological change in the microbiota. In this review, we provide clear and concise discussions on the human gut microbiota, methods of identifying gut microbiota and its functionality, liver diseases that are affected by the gut microbiota, including novel associations under research, and provide current evidence on the modulation of gut microbiota and its effects on specific liver disease conditions.
Collapse
Affiliation(s)
- Cyriac Abby Philips
- The Liver Unit, Monarch Liver Lab and Division of Gastroenterology, Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| | - Philip Augustine
- The Liver Unit, Monarch Liver Lab and Division of Gastroenterology, Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| | - Praveen Kumar Yerol
- Department of Gastroenterology, State Government Medical College, Thrissur, Kerala, India
| | | | - Rizwan Ahamed
- The Liver Unit, Monarch Liver Lab and Division of Gastroenterology, Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| | - Sasidharan Rajesh
- The Liver Unit, Monarch Liver Lab and Division of Gastroenterology, Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| | - Tom George
- The Liver Unit, Monarch Liver Lab and Division of Gastroenterology, Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| | - Sandeep Kumbar
- The Liver Unit, Monarch Liver Lab and Division of Gastroenterology, Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| |
Collapse
|
458
|
Dinleyici M, Pérez-Brocal V, Arslanoglu S, Aydemir O, Ozumut SS, Tekin N, Vandenplas Y, Moya A, Dinleyici EC. Human milk mycobiota composition: relationship with gestational age, delivery mode, and birth weight. Benef Microbes 2020; 11:151-162. [PMID: 31990220 DOI: 10.3920/bm2019.0158] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Intestinal and human milk microbiota studies during infancy have shown variations according to geographical location, delivery mode, gestational age, and mother-related factors during pregnancy. In this study, we performed metagenomic mycobiota analyses of 44 transient and mature human milk among five different groups: mothers of normal spontaneous delivery-term (NS-T), caesarean delivery-term (CS-T), premature (PT), small for gestational age (SGA), and large for gestational age (LGA) infants. Fungi were detected in 80 out of the 88 samples. Regarding the number of observed fungal species, the NS-T group was more homogeneous (less variable) comparing the other groups (P<0.05). In the transient human milk samples, the most abundant species were Saccharomyces cerevisiae (33.3%) and Aspergillus glaucus (27.4%). While A. glaucus (33.7%) was second most abundant species in mature milk, S. cerevisiae disappeared (P<0.01) and Penicillium rubens became the most abundant species (35.5%) (P<0.05). Among the NS-T group, the most abundant species was Malassezia globosa in both transient and mature milk. In contrast, S. cerevisiae was the most abundant species in transient human milk (45.0%) in the CS-T group, but it disappeared in mature milk (P<0.01). In transient milk, M. globosa was only represented 6.0-9.0% of taxa in the PT, SGA, and LGA groups (P<0.05). In transient and mature milk in the PT, SGA and LGA groups, the most abundant species were A. glaucus and P. rubens. In mature milk samples, P. rubens is more abundant in CS-T group, PT group and LGA group, than the NS-T groups (P<0.05 for all). Although fungi constitute only a very small part of the human milk microbiome, we observed some changes that the human milk mycobiota composition varies in caesarean delivery, premature, SGA and LGA groups, comparing the normal spontaneous delivery, as well as differences between transient and mature human milk.
Collapse
Affiliation(s)
- M Dinleyici
- Eskisehir Osmangazi University Faculty of Medicine, Department of Pediatrics, Eskisehir 26480, Turkey
- Eskisehir Osmangazi University Faculty of Medicine, Department of Social Pediatrics, Eskisehir 26480, Turkey
| | - V Pérez-Brocal
- Department of Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health), Valencia, Spain
- CIBER in Epidemiology and Public Health (CIBEResp), Madrid, Spain
| | - S Arslanoglu
- Division of Neonatology, Department of Pediatrics, Istanbul Medeniyet University Faculty of Medicine, Istanbul, Turkey
| | - O Aydemir
- Eskisehir Osmangazi University Faculty of Medicine, Department of Pediatrics, Eskisehir 26480, Turkey
- Eskisehir Osmangazi University Faculty of Medicine, Division of Neonatology, Eskisehir 26480, Turkey
| | - S Sevuk Ozumut
- Division of Neonatology, Department of Pediatrics, Istanbul Medeniyet University Faculty of Medicine, Istanbul, Turkey
| | - N Tekin
- Eskisehir Osmangazi University Faculty of Medicine, Department of Pediatrics, Eskisehir 26480, Turkey
- Eskisehir Osmangazi University Faculty of Medicine, Division of Neonatology, Eskisehir 26480, Turkey
| | - Y Vandenplas
- UZ Brussel, Vrije Universiteit Brussel, Department of Pediatrics, Brussel, Belgium
| | - A Moya
- Department of Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health), Valencia, Spain
- CIBER in Epidemiology and Public Health (CIBEResp), Madrid, Spain
- Institute for Integrative Systems Biology (I2SysBio), The University of Valencia and The Spanish National Research Council (CSIC-UVEG), Valencia, Spain
| | - E C Dinleyici
- Eskisehir Osmangazi University Faculty of Medicine, Department of Pediatrics, Eskisehir 26480, Turkey
| |
Collapse
|
459
|
Specific Eco-evolutionary Contexts in the Mouse Gut Reveal Escherichia coli Metabolic Versatility. Curr Biol 2020; 30:1049-1062.e7. [PMID: 32142697 DOI: 10.1016/j.cub.2020.01.050] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/21/2019] [Accepted: 01/15/2020] [Indexed: 02/08/2023]
Abstract
Members of the gut microbiota are thought to experience strong competition for nutrients. However, how such competition shapes their evolutionary dynamics and depends on intra- and interspecies interactions is poorly understood. Here, we test the hypothesis that Escherichia coli evolution in the mouse gut is more predictable across hosts in the absence of interspecies competition than in the presence of other microbial species. In support, we observed that lrp, a gene encoding a global regulator of amino acid metabolism, was repeatedly selected in germ-free mice 2 weeks after mono-colonization by this bacterium. We established that this specific genetic adaptation increased E. coli's ability to compete for amino acids, and analysis of gut metabolites identified serine and threonine as the metabolites preferentially consumed by E. coli in the mono-colonized mouse gut. Preference for serine consumption was further supported by testing a set of mutants that showed loss of advantage of an lrp mutant impaired in serine metabolism in vitro and in vivo. Remarkably, the presence of a single additional member of the microbiota, Blautia coccoides, was sufficient to alter the gut metabolome and, consequently, the evolutionary path of E. coli. In this environment, the fitness advantage of the lrp mutant bacteria is lost, and mutations in genes involved in anaerobic respiration were selected instead, recapitulating the eco-evolutionary context from mice with a complex microbiota. Together, these results highlight the metabolic plasticity and evolutionary versatility of E. coli, tailored to the specific ecology it experiences in the gut.
Collapse
|
460
|
Gupta V, Kumar R, Sood U, Singhvi N. Reconciling Hygiene and Cleanliness: A New Perspective from Human Microbiome. Indian J Microbiol 2020; 60:37-44. [PMID: 32089572 PMCID: PMC7000587 DOI: 10.1007/s12088-019-00839-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/03/2019] [Indexed: 12/12/2022] Open
Abstract
The term hygiene is deeply rooted with the concept of maintaining sound health and alertness towards cleanliness, while "hygiene hypothesis" depicts the protective role of microbial community exposure in development of early immunity and initial allergic and aesthetic reactions. The tug-of-war has now been pushed toward the literal term "hygiene" over the "hygiene hypothesis" and has continued with disinfection of all microbial loads from the related environments to avoid infections in humans. With the advancement in the microbiome studies, it became clear that humans possess warm, and significant relationships with diverse microbial community. With this opinion article, we have emphasized on the importance of hygiene hypothesis in immunological responses. We also propose the individual/targeted hygiene instead of application of unanimous hygiene hypothesis. This review also elaborates the common practices that should be employed to maintain hygiene along with the balanced microbiome.
Collapse
Affiliation(s)
- Vipin Gupta
- PhiXGen Private Limited, Gurugram, Haryana 122001 India
- Department of Zoology, University of Delhi, Delhi, Delhi 110007 India
| | - Roshan Kumar
- P.G. Department of Zoology, Magadh University, Bodh-Gaya, Bihar 824231 India
| | - Utkarsh Sood
- The Energy and Resources Institute, Darbari Seth Block, IHC Complex, Lodhi Road, New Delhi, 110003 India
| | - Nirjara Singhvi
- Department of Zoology, University of Delhi, Delhi, Delhi 110007 India
| |
Collapse
|
461
|
Vandana UK, Barlaskar NH, Gulzar ABM, Laskar IH, Kumar D, Paul P, Pandey P, Mazumder PB. Linking gut microbiota with the human diseases. Bioinformation 2020; 16:196-208. [PMID: 32405173 PMCID: PMC7196170 DOI: 10.6026/97320630016196] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
The human gut is rich in microbes. Therefore, it is of interest to document data to link known human diseases with the gut microbiota. Various factors like hormones, metabolites and dietary habitats are responsible for shaping the microbiota of the gut. Imbalance in the gut microbiota is responsible for the pathogenesis of various disease types including rheumatoid arthritis, different types of cancer, diabetes mellitus, obesity, and cardiovascular disease. We report a review of known data for the correction of dysbiosis (imbalance in microbe population) towards improved human health.
Collapse
Affiliation(s)
| | | | | | | | - Diwakar Kumar
- Department of Microbiology, Assam University, Silchar, Assam, India
| | - Prosenjit Paul
- Department of Biotechnology, Assam University, Silchar, Assam, India
| | - Piyush Pandey
- Department of Microbiology, Assam University, Silchar, Assam, India
| | | |
Collapse
|
462
|
Doumatey AP, Adeyemo A, Zhou J, Lei L, Adebamowo SN, Adebamowo C, Rotimi CN. Gut Microbiome Profiles Are Associated With Type 2 Diabetes in Urban Africans. Front Cell Infect Microbiol 2020; 10:63. [PMID: 32158702 PMCID: PMC7052266 DOI: 10.3389/fcimb.2020.00063] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/07/2020] [Indexed: 01/04/2023] Open
Abstract
Gut dysbiosis has been associated with several disease outcomes including diabetes in human populations. Currently, there are no studies of the gut microbiome composition in relation to type 2 diabetes (T2D) in Africans. Here, we describe the profile of the gut microbiome in non-diabetic adults (controls) and investigate the association between gut microbiota and T2D in urban West Africans. Gut microbiota composition was determined in 291 Nigerians (98 cases, 193 controls) using fecal 16S V4 rRNA gene sequencing done on the Illumina MiSeq platform. Data analysis of operational taxonomic units (OTU) was conducted to describe microbiome composition and identify differences between T2D and controls. The most abundant phyla were Firmicutes, Actinobacteria, and Bacteroidetes. Clostridiaceae, and Peptostreptococcaceaea were significantly lower in cases than controls (p < 0.001). Feature selection analysis identified a panel of 18 OTUs enriched in cases that included Desulfovibrio piger, Prevotella, Peptostreptococcus, and Eubacterium. A panel of 17 OTUs that was enriched in the controls included Collinsella, Ruminococcus lactaris, Anaerostipes, and Clostridium. OTUs with strain-level annotation showing the largest fold-change included Cellulosilyticum ruminicola (log2FC = −3.1; p = 4.2 × 10−5), Clostridium paraputrificum (log2FC = −2.5; p = 0.005), and Clostridium butyricum (log2FC = −1.76; p = 0.01), all lower in cases. These findings are notable because supplementation with Clostridium butyricum and Desulfovibrio piger has been shown to improve hyperglycemia and reduce insulin resistance in murine models. This first investigation of gut microbiome and diabetes in urban Africans shows that T2D is associated with compositional changes in gut microbiota highlighting the possibility of developing strategies to improve glucose control by modifying bacterial composition in the gut.
Collapse
Affiliation(s)
- Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jie Zhou
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Lin Lei
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sally N Adebamowo
- Department of Epidemiology and Public Health and Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States.,Center for Bioethics and Research, Ibadan, Nigeria
| | - Clement Adebamowo
- Department of Epidemiology and Public Health and Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States.,Center for Bioethics and Research, Ibadan, Nigeria.,Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
463
|
Lokmer A, Aflalo S, Amougou N, Lafosse S, Froment A, Tabe FE, Poyet M, Groussin M, Said-Mohamed R, Ségurel L. Response of the human gut and saliva microbiome to urbanization in Cameroon. Sci Rep 2020; 10:2856. [PMID: 32071424 PMCID: PMC7028744 DOI: 10.1038/s41598-020-59849-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/05/2020] [Indexed: 12/22/2022] Open
Abstract
Urban populations from highly industrialized countries are characterized by a lower gut bacterial diversity as well as by changes in composition compared to rural populations from less industrialized countries. To unveil the mechanisms and factors leading to this diversity loss, it is necessary to identify the factors associated with urbanization-induced shifts at a smaller geographical scale, especially in less industrialized countries. To do so, we investigated potential associations between a variety of dietary, medical, parasitological and socio-cultural factors and the gut and saliva microbiomes of 147 individuals from three populations along an urbanization gradient in Cameroon. We found that the presence of Entamoeba sp., a commensal gut protozoan, followed by stool consistency, were major determinants of the gut microbiome diversity and composition. Interestingly, urban individuals have retained most of their gut eukaryotic and bacterial diversity despite significant changes in diet compared to the rural areas, suggesting that the loss of bacterial microbiome diversity observed in industrialized areas is likely associated with medication. Finally, we observed a weak positive correlation between the gut and the saliva microbiome diversity and composition, even though the saliva microbiome is mainly shaped by habitat-related factors.
Collapse
Affiliation(s)
- Ana Lokmer
- UMR7206 Eco-anthropologie, CNRS - MNHN - Université de Paris, Paris, France.
| | - Sophie Aflalo
- UMR7206 Eco-anthropologie, CNRS - MNHN - Université de Paris, Paris, France
| | - Norbert Amougou
- UMR7206 Eco-anthropologie, CNRS - MNHN - Université de Paris, Paris, France
| | - Sophie Lafosse
- UMR7206 Eco-anthropologie, CNRS - MNHN - Université de Paris, Paris, France
| | - Alain Froment
- UMR7206 Eco-anthropologie, CNRS - MNHN - Université de Paris, Paris, France
| | - Francis Ekwin Tabe
- Faculté de Médecine et des Sciences Biomédicales - Université Yaoundé 1, Yaoundé, Cameroun
| | - Mathilde Poyet
- Department of Biological Engineering/Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mathieu Groussin
- Department of Biological Engineering/Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rihlat Said-Mohamed
- SAMRC/WITS Developmental Pathways for Health Research Unit, Department of Paediatrics, School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Laure Ségurel
- UMR7206 Eco-anthropologie, CNRS - MNHN - Université de Paris, Paris, France.
| |
Collapse
|
464
|
Abstract
We are in the midst of “the microbiome revolution”—not a day goes by without some new revelation on the potential role of the gut microbiome in some disease or disorder. From an ever-increasing recognition of the many roles of the gut microbiome in health and disease comes the expectation that its modulation could treat or prevent these very same diseases. A variety of interventions could, at least in theory, be employed to alter the composition or functional capacity of the microbiome, ranging from diet to fecal microbiota transplantation (FMT). For some, such as antibiotics, prebiotics, and probiotics, an extensive, albeit far from consistent, literature already exists; for others, such as other dietary supplements and FMT, high-quality clinical studies are still relatively few in number. Not surprisingly, researchers have turned to the microbiome itself as a source for new entities that could be used therapeutically to manipulate the microbiome; for example, some probiotic strains currently in use were sourced from the gastrointestinal tract of healthy humans. From all of the extant studies of interventions targeted at the gut microbiome, a number of important themes have emerged. First, with relatively few exceptions, we are still a long way from a precise definition of the role of the gut microbiome in many of the diseases where a disturbed microbiome has been described—association does not prove causation. Second, while animal models can provide fascinating insights into microbiota–host interactions, they rarely recapitulate the complete human phenotype. Third, studies of several interventions have been difficult to interpret because of variations in study population, test product, and outcome measures, not to mention limitations in study design. The goal of microbiome modulation is a laudable one, but we need to define our targets, refine our interventions, and agree on outcomes.
Collapse
Affiliation(s)
- Eamonn M M Quigley
- Lynda K and David M Underwood Center for Digestive Disorders, Division of Gastroenterology and Hepatology, Houston Methodist Hospital, Houston, Texas, 77030, USA
| | - Prianka Gajula
- Department of Medicine, Houston Methodist Hospital, Houston, Texas, 77030, USA
| |
Collapse
|
465
|
The human microbiome in sickness and in health. Rev Clin Esp 2020; 221:233-240. [PMID: 33998505 DOI: 10.1016/j.rceng.2019.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 07/08/2019] [Indexed: 01/22/2023]
Abstract
The study of the human microbiome has led to an exceptional increase in the current understanding of the importance of microbiota for health throughout all stages of life. Human microbial colonization occurs in the skin, genitourinary system and, mainly, in the oral cavity and intestinal tract. In these locations, the human microbiota establishes a symbiotic relationship with the host and helps maintain physiological homeostasis. Lifestyle, age, diet and use of antibiotics are the main regulators of the composition and functionality of human microbiota. Recent studies have indicated the reduction in microbial diversity as one of the contributors to the development of diseases. In addition to phylogenetic diversity studies, further metagenomic studies are needed at the functional level of the human microbiome to improve our understanding of its involvement in human health.
Collapse
|
466
|
Abo H, Chassaing B, Harusato A, Quiros M, Brazil JC, Ngo VL, Viennois E, Merlin D, Gewirtz AT, Nusrat A, Denning TL. Erythroid differentiation regulator-1 induced by microbiota in early life drives intestinal stem cell proliferation and regeneration. Nat Commun 2020; 11:513. [PMID: 31980634 PMCID: PMC6981263 DOI: 10.1038/s41467-019-14258-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 12/22/2019] [Indexed: 01/07/2023] Open
Abstract
Gut microbiota and their metabolites are instrumental in regulating intestinal homeostasis. However, early-life microbiota associated influences on intestinal development remain incompletely understood. Here we demonstrate that co-housing of germ-free (GF) mice with specific-pathogen free (SPF) mice at weaning (exGF) results in altered intestinal gene expression. Our results reveal that one highly differentially expressed gene, erythroid differentiation regulator-1 (Erdr1), is induced during development in SPF but not GF or exGF mice and localizes to Lgr5+ stem cells and transit amplifying (TA) cells. Erdr1 functions to induce Wnt signaling in epithelial cells, increase Lgr5+ stem cell expansion, and promote intestinal organoid growth. Additionally, Erdr1 accelerates scratch-wound closure in vitro, increases Lgr5+ intestinal stem cell regeneration following radiation-induced injury in vivo, and enhances recovery from dextran sodium sulfate (DSS)-induced colonic damage. Collectively, our findings indicate that early-life microbiota controls Erdr1-mediated intestinal epithelial proliferation and regeneration in response to mucosal damage. Gut microbiota and their metabolites regulate homeostasis of the intestine, but their effects on intestine development are unclear. Here the authors use RNAseq and germ free mice to show that intestinal microbiota promote the expression of Erdr1, which increases Lgr5+ intestinal stem cell number and activity.
Collapse
Affiliation(s)
- Hirohito Abo
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Benoit Chassaing
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA.,Neuroscience Institute and Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA.,INSERM, U1016, Paris, France.,Université de Paris, Paris, France
| | - Akihito Harusato
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Miguel Quiros
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jennifer C Brazil
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Vu L Ngo
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Emilie Viennois
- Center for Diagnostics and Therapeutics, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Didier Merlin
- Center for Diagnostics and Therapeutics, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.,Atlanta Veterans Affairs Medical Center, Decatur, GA, 30033, USA
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Timothy L Denning
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA.
| |
Collapse
|
467
|
Cross JH, Jarjou O, Mohammed NI, Rayment Gomez S, Touray BJB, Prentice AM, Cerami C. Early postnatal hypoferremia in low birthweight and preterm babies: A prospective cohort study in hospital-delivered Gambian neonates. EBioMedicine 2020; 52:102613. [PMID: 31981986 PMCID: PMC6992934 DOI: 10.1016/j.ebiom.2019.102613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/29/2019] [Accepted: 12/17/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Neonates, particularly those born preterm (PTB) and with low birthweight (LBW), are especially susceptible to bacterial and fungal infections that cause an estimated 225,000 deaths annually. Iron is a vital nutrient for the most common organisms causing septicaemia. Full-term babies elicit an immediate postnatal hypoferremia assumed to have evolved as an innate defence. We tested whether PTB and LBW babies are capable of the same response. METHODS We conducted an observational study of 152 babies who were either PTB (born ≥32 to <37 weeks gestational age) and/or LBW (<2500 g) (PTB/LBW) and 278 term, normal-weight babies (FTB/NBW). Blood was sampled from the umbilical cord vein and artery, and matched venous blood samples were taken from all neonates between 6-24 h after delivery. We measured haematological, iron and inflammatory markers. FINDINGS In both PTB/LBW and FTB/NBW babies, serum iron decreased 3-fold within 12 h of delivery compared to umbilical blood (7·5 ± 4·5 vs 23·3 ± 7·1 ng/ml, P < 0·001, n = 425). Transferrin saturation showed a similar decline with a consequent increase in unsaturated iron-binding capacity. C-reactive protein levels increased over 10-fold (P < 0·001) and hepcidin levels doubled (P < 0·001). There was no difference in any of these responses between PTB/LBW and FTB/NBW babies. INTERPRETATION Premature or low birthweight babies are able to mount a very rapid hypoferremia that is indistinguishable from that in normal term babies. The data suggest that this is a hepcidin-mediated response triggered by acute inflammation at birth, and likely to have evolved as an innate immune response against bacterial and fungal septicaemia. TRIAL REGISTRATION clinicaltrials.gov (NCT03353051). Registration date: November 27, 2017. FUNDING Bill & Melinda Gates Foundation (OPP1152353).
Collapse
Affiliation(s)
- James H Cross
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Atlantic Boulevard, Fajara, P.O. Box 273, Banjul, The Gambia
| | - Ousman Jarjou
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Atlantic Boulevard, Fajara, P.O. Box 273, Banjul, The Gambia
| | - Nuredin Ibrahim Mohammed
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Atlantic Boulevard, Fajara, P.O. Box 273, Banjul, The Gambia
| | | | - Bubacarr J B Touray
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Atlantic Boulevard, Fajara, P.O. Box 273, Banjul, The Gambia
| | - Andrew M Prentice
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Atlantic Boulevard, Fajara, P.O. Box 273, Banjul, The Gambia
| | - Carla Cerami
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Atlantic Boulevard, Fajara, P.O. Box 273, Banjul, The Gambia.
| |
Collapse
|
468
|
Underwood MA, Mukhopadhyay S, Lakshminrusimha S, Bevins CL. Neonatal intestinal dysbiosis. J Perinatol 2020; 40:1597-1608. [PMID: 32968220 PMCID: PMC7509828 DOI: 10.1038/s41372-020-00829-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/17/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022]
Abstract
The initial colonization of the neonatal intestinal tract is influenced by delivery mode, feeding, the maternal microbiota, and a host of environmental factors. After birth, the composition of the infant's microbiota undergoes a series of significant changes particularly in the first weeks and months of life ultimately developing into a more stable and diverse adult-like population in childhood. Intestinal dysbiosis is an alteration in the intestinal microbiota associated with disease and appears to be common in neonates. The consequences of intestinal dysbiosis are uncertain, but strong circumstantial evidence and limited confirmations of causality suggest that dysbiosis early in life can influence the health of the infant acutely, as well as contribute to disease susceptibility later in life.
Collapse
Affiliation(s)
- Mark A. Underwood
- grid.27860.3b0000 0004 1936 9684Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA USA
| | - Sagori Mukhopadhyay
- grid.25879.310000 0004 1936 8972Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA
| | - Satyan Lakshminrusimha
- grid.27860.3b0000 0004 1936 9684Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA USA
| | - Charles L. Bevins
- grid.27860.3b0000 0004 1936 9684Department of Medical Microbiology and Immunology, UC Davis School of Medicine, Davis, CA USA
| |
Collapse
|
469
|
Manipulating resident microbiota to enhance regulatory immune function to treat inflammatory bowel diseases. J Gastroenterol 2020; 55:4-14. [PMID: 31482438 PMCID: PMC6942586 DOI: 10.1007/s00535-019-01618-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 08/20/2019] [Indexed: 02/04/2023]
Abstract
Altered intestinal microbial composition (dysbiosis) and metabolic products activate aggressive mucosal immune responses that mediate inflammatory bowel diseases (IBD). This dysbiosis impairs the function of regulatory immune cells, which normally promote mucosal homeostasis. Normalizing and maintaining regulatory immune cell function by correcting dysbiosis provides a promising approach to treat IBD patients. However, existing microbe-targeted therapies, including antibiotics, prebiotics, probiotics, and fecal microbial transplantation, provide variable outcomes that are not optimal for current clinical application. This review discusses recent progress in understanding the dysbiosis of IBD and the basis for therapeutic restoration of homeostatic immune function by manipulating an individual patient's microbiota composition and function. We believe that identifying more precise therapeutic targets and developing appropriate rapid diagnostic tools will guide more effective and safer microbe-based induction and maintenance treatments for IBD patients that can be applied in a personalized manner.
Collapse
|
470
|
Li M, Shao D, Zhou J, Gu J, Qin J, Chen W, Wei W. Signatures within esophageal microbiota with progression of esophageal squamous cell carcinoma. Chin J Cancer Res 2020; 32:755-767. [PMID: 33446998 PMCID: PMC7797230 DOI: 10.21147/j.issn.1000-9604.2020.06.09] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective Esophageal squamous cell carcinoma (ESCC) is one of the dominant malignances worldwide, but currently there is less focus on the microbiota with ESCC and its precancerous lesions. Methods Paired esophageal biopsy and swab specimens were obtained from 236 participants in Linzhou, China. Data from 16S ribosomal RNA gene sequencing were processed using quantitative insights into microbial ecology (QIIME2) and R Studio to evaluate differences. The Wilcoxon rank sum test and Kruskal-Wallis rank sum test were used to compare diversity and characteristic genera by specimens and participant groups. Ordinal logistic regression model was used to build microbiol prediction model. Results Microbial diversity was similar between biopsy and swab specimens, including operational taxonomic unit (OTU) numbers and Shannon index. There were variations and similarities of esophageal microbiota among different pathological characteristics of ESCC. Top 10 relative abundance genera in all groups include Streptococcus, Prevotella, Veillonella, Actinobacillus, Haemophilus, Neisseria, Alloprevotella, Rothia, Gemella and Porphyromonas. Genus Streptococcus, Haemophilus, Neisseria and Porphyromonas showed significantly difference in disease groups when compared to normal control, whereas Streptococcus showed an increasing tendency with the progression of ESCC and others showed a decreasing tendency. About models based on all combinations of characteristic genera, only taken Streptococcus and Neisseria into model, the prediction performance was the ideal one, of which the area under the curve (AUC) was 0.738.
Conclusions Esophageal biopsy and swab specimens could yield similar microbial characterization. The combination of Streptococcus and Neisseria has the potential to predict the progression of ESCC, which is needed to confirm by large-scale, prospective cohort studies.
Collapse
Affiliation(s)
- Minjuan Li
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dantong Shao
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jiachen Zhou
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Jianhua Gu
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Junjie Qin
- Promegene Translational Research Institute, Shenzhen 518000, China
| | - Wen Chen
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wenqiang Wei
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
471
|
Is the Newborn Microbiome Disrupted by Routine Newborn Suctioning? An Exploratory Approach for Policy Development. J Perinat Neonatal Nurs 2020; 34:231-238. [PMID: 32697543 DOI: 10.1097/jpn.0000000000000499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
An invasive procedure commonly used in the perinatal setting is newborn suctioning at birth. Routine newborn suctioning, without indication, is not recommended by the American Academy of Pediatrics, the American Heart Association, and the European Resuscitation Council for the newborn with spontaneous respirations, adequate crying, and good muscle tone. Whether to suction a newborn is a difficult practice decision made daily by the perinatal nurse. A primary element of this practice decision is the consideration that newborn suctioning has the potential to affect health outcomes. Furthermore, routine newborn suctioning may be disrupting the newborn microbiome by removing commensal bacteria and potentially, negatively affecting newborn health. This article will explore the connection between the practice of routine newborn suctioning in the perinatal setting and the potential to disrupt the newborn microbiome. The methods employed and reported in this article consisted of review of literature and review of clinical guidelines and a descriptive study to determine the scope of practice of newborn suctioning. The premise of whether the practice of routine newborn suctioning at birth may be disrupting the newborn microbiome will also be considered. Finally, developing a microbiome-centric perspective will be explored.
Collapse
|
472
|
van Tilburg Bernardes E, Gutierrez MW, Arrieta MC. The Fungal Microbiome and Asthma. Front Cell Infect Microbiol 2020. [PMID: 33324573 DOI: 10.3389/fcimb.2020.583418/full] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Asthma is a group of inflammatory conditions that compromises the airways of a continuously increasing number of people around the globe. Its complex etiology comprises both genetic and environmental aspects, with the intestinal and lung microbiomes emerging as newly implicated factors that can drive and aggravate asthma. Longitudinal infant cohort studies combined with mechanistic studies in animal models have identified microbial signatures causally associated with subsequent asthma risk. The recent inclusion of fungi in human microbiome surveys has revealed that microbiome signatures associated with asthma risk are not limited to bacteria, and that fungi are also implicated in asthma development in susceptible individuals. In this review, we examine the unique properties of human-associated and environmental fungi, which confer them the ability to influence immune development and allergic responses. The important contribution of fungi to asthma development and exacerbations prompts for their inclusion in current and future asthma studies in humans and animal models.
Collapse
Affiliation(s)
- Erik van Tilburg Bernardes
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Mackenzie W Gutierrez
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
473
|
Health risk behaviours and allostatic load: A systematic review. Neurosci Biobehav Rev 2020; 108:694-711. [DOI: 10.1016/j.neubiorev.2019.12.020] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/05/2019] [Accepted: 12/11/2019] [Indexed: 12/26/2022]
|
474
|
Laboratory Analysis Techniques for the Perinatal Microbiome: Implications for Studies of Probiotic Interventions. J Perinat Neonatal Nurs 2020; 34:239-250. [PMID: 32697544 PMCID: PMC9528808 DOI: 10.1097/jpn.0000000000000496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The microbiome is composed of many organisms and is impacted by an intricate exchange between genetics and environmental factors. The perinatal microbiome influences both the developing fetus and the pregnant person. The purpose of this article is to describe the tests that are currently available for laboratory analysis of the perinatal microbiome in relationship to probiotic interventions. This article focuses on the bacterial component of the microbiome. Although adverse outcomes associated with the perinatal microbiome have been studied, a comprehensive understanding of the physiologic perinatal microbiome is still emerging. Early efforts to influence the perinatal microbiome through probiotics are currently under investigation. Unique terminology is defined, and the microbial composition of perinatal microbiota is summarized. The outcomes of studies of antenatal probiotics are summarized. Microbiome testing and analysis are defined and compared. Implications for perinatal care and probiotics research are presented.
Collapse
|
475
|
Sambucci M, Gargano F, Guerrera G, Battistini L, Borsellino G. One, No One, and One Hundred Thousand: T Regulatory Cells' Multiple Identities in Neuroimmunity. Front Immunol 2019; 10:2947. [PMID: 31956323 PMCID: PMC6955595 DOI: 10.3389/fimmu.2019.02947] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022] Open
Abstract
As the Nobel laureate Luigi Pirandello wrote in his novels, identities can be evanescent. Although a quarter of a century has passed since regulatory T cells (Treg) were first described, new studies continue to reveal surprising and contradictory features of this lymphocyte subset. Treg cells are the core of the immunological workforce engaged in the restraint of autoimmune or inflammatory reactions, and their characterization has revealed substantial heterogeneity and complexity in the phenotype and gene expression profiles, proving them to be a most versatile and adaptive cell type, as exemplified by their plasticity in fine-tuning immune responses. Defects in Treg function are associated with several autoimmune diseases, including multiple sclerosis, which is caused by an inappropriate immune reaction toward brain components; conversely, the beneficial effects of immunomodulating therapies on disease progression have been shown to partly act upon the biology of these cells. Both in animals and in humans the pool of circulating Treg cells is a mixture of natural (nTregs) and peripherally-induced Treg (pTregs). Particularly in humans, circulating Treg cells can be phenotypically subdivided into different subpopulations, which so far are not well-characterized, particularly in the context of autoimmunity. Recently, Treg cells have been rediscovered as mediators of tissue healing, and have also shown to be involved in organ homeostasis. Moreover, stability of the Treg lineage has recently been addressed by several conflicting reports, and immune-suppressive abilities of these cells have been shown to be dynamically regulated, particularly in inflammatory conditions, adding further levels of complexity to the study of this cell subset. Finally, Treg cells exert their suppressive function through different mechanisms, some of which—such as their ectoenzymatic activity—are particularly relevant in CNS autoimmunity. Here, we will review the phenotypically and functionally discernible Treg cell subpopulations in health and in multiple sclerosis, touching also upon the effects on this cell type of immunomodulatory drugs used for the treatment of this disease.
Collapse
Affiliation(s)
- Manolo Sambucci
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | | | | | - Luca Battistini
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | | |
Collapse
|
476
|
Zhang X, Browman G, Siu W, Basen-Engquist KM, Hanash SM, Hoffman KL, Okhuysen PC, Scheet P, Petrosino JF, Kopetz S, Daniel CR. The BE GONE trial study protocol: a randomized crossover dietary intervention of dry beans targeting the gut microbiome of overweight and obese patients with a history of colorectal polyps or cancer. BMC Cancer 2019; 19:1233. [PMID: 31852462 PMCID: PMC6921460 DOI: 10.1186/s12885-019-6400-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mouse and human studies support the promise of dry beans to improve metabolic health and to lower cancer risk. In overweight/obese patients with a history of colorectal polyps or cancer, the Beans to Enrich the Gut microbiome vs. Obesity's Negative Effects (BE GONE) trial will test whether and how an increase in the consumption of pre-cooked, canned dry beans within the context of usual diet and lifestyle can enhance the gut landscape to improve metabolic health and reduce cancer risk. METHODS/DESIGN This randomized crossover trial is designed to characterize changes in (1) host markers spanning lipid metabolism, inflammation, and obesity-related cancer risk; (2) compositional and functional profiles of the fecal microbiome; and (3) host and microbial metabolites. With each subject serving as their own control, the trial will compare the participant's usual diet with (intervention) and without (control) dry beans. Canned, pre-cooked dry beans are provided to participants and the usual diet continually assessed and monitored. Following a 4-week run-in and equilibration period, each participant provides a total of 5 fasting blood and 6 stool samples over a total period of 16 weeks. The intervention consists of a 2-week ramp-up of dry bean intake to 1 cup/d, which is then continued for an additional 6 weeks. Intra- and inter-individual outcomes are assessed across each crossover period with consideration of the joint or modifying effects of the usual diet and baseline microbiome. DISCUSSION The BE GONE trial is evaluating a scalable dietary prevention strategy targeting the gut microbiome of high-risk patients to mitigate the metabolic and inflammatory effects of adiposity that influence colorectal cancer risk, recurrence, and survival. The overarching scientific goal is to further elucidate interactions between diet, the gut microbiome, and host metabolism. Improved understanding of the diet-microbiota interplay and effective means to target these relationships will be key to the future of clinical and public health approaches to cancer and other major diet- and obesity-related diseases. TRIAL REGISTRATION This protocol is registered with the U.S. National Institutes of Health trial registry, ClinicalTrials.gov, under the identifier NCT02843425. First posted July 25, 2016; last verified January 25, 2019.
Collapse
Affiliation(s)
- Xiaotao Zhang
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1340, Houston, TX, TX 77030, USA
- Department of Medicine, Epidemiology and Population Science, Baylor College of Medicine, Houston, TX, USA
| | - Gladys Browman
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1340, Houston, TX, TX 77030, USA
| | - Wesley Siu
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1340, Houston, TX, TX 77030, USA
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Karen M Basen-Engquist
- Department of Behavioral Science, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristi L Hoffman
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Pablo C Okhuysen
- Department of Infectious Diseases, Infection Control, and Employee Health, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Scheet
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1340, Houston, TX, TX 77030, USA
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carrie R Daniel
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1340, Houston, TX, TX 77030, USA.
| |
Collapse
|
477
|
Singh R, Haque MM, Mande SS. Lifestyle-Induced Microbial Gradients: An Indian Perspective. Front Microbiol 2019; 10:2874. [PMID: 31921052 PMCID: PMC6928055 DOI: 10.3389/fmicb.2019.02874] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 11/28/2019] [Indexed: 02/06/2023] Open
Abstract
Introduction: Urbanization is a globally pervasive trend. Although urban settings provide better access to infrastructure and opportunities, urban lifestyles have certain negative consequences on human health. A number of recent studies have found interesting associations between the structure of human gut microbiota and the prevalence of metabolic conditions characterizing urban populations. The present study attempts to expand the footprint of these investigations to an Indian context. The objectives include elucidating specific patterns and gradients based on resident habitat and lifestyles (i.e., tribal and urban) that characterize gut microbial communities. Methods: Available 16S rRNA sequence datasets corresponding to the gut microbiota of urban and tribal populations from multiple regions of India have been rigorously compared. This analysis was carried out to understand the overall community structure, resident taxa, and their (inferred) functional components as well as their correlations with available meta-information. Results: The gut microbiota of urban and tribal communities are observed to have characteristically different signatures with respect to diversity as well as taxonomic and functional composition. Primarily, the gut microbiota in tribal communities is found to harbor significantly higher species diversity and richness as compared to that in urban populations. In spite of geographical segregation and diet-related differences, gut microbial diversity was not found to differ significantly between tribal groups. Furthermore, while the taxonomic profiles of different tribal communities cluster together irrespective of their geographic location, enterotype analysis indicates that samples from urban communities form two distinct clusters. Taxonomic analysis of samples in one of these clusters reveals the presence of microbes that are common to both urban and tribal cohorts, indicating a probable transient evolutionary state. Prevotella, previously reported to be the dominant genus resident in Indian gut microbiota, is found to have distinct OTUs and strain-specific oligotypes characterizing resident habitats and diet patterns. Certain interesting associations between microbial abundances and specific metadata have also been observed. Overall, urban lifestyle and diet appear to impact the structure and function of gut microbial communities, and the results of this study provide further evidence of this likely detrimental association. Conclusion: This study attempts to analyze, in an Indian context, the impact of urbanization on the human gut microbiota. Overall, the analysis elucidates interesting taxonomic and functional signatures characterizing the evolutionary transition in gut microbiota from tribal to urban.
Collapse
Affiliation(s)
- Rashmi Singh
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services, Pune, India
| | | | - Sharmila S Mande
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services, Pune, India
| |
Collapse
|
478
|
Peterson SN, Bradley LM, Ronai ZA. The gut microbiome: an unexpected player in cancer immunity. Curr Opin Neurobiol 2019; 62:48-52. [PMID: 31816571 DOI: 10.1016/j.conb.2019.09.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 09/24/2019] [Indexed: 12/11/2022]
Abstract
Numerous independent studies link gut microbiota composition and disease and imply a causal role of select commensal microbes in disease etiology. In the gut, commensal microbiota or pathobionts secrete metabolites that underlie pathological conditions, often impacting proximal tissues and gaining access to the bloodstream. Here we focus on extrinsic and intrinsic factors affecting composition of gut microbiota and their impact on the immune system, as key drivers of anti-tumor immunity. In discussing exciting advances relevant to microbiome-tumor interaction, we note existing knowledge gaps that need to be filled to advance basic and clinical research initiatives.
Collapse
Affiliation(s)
- Scott N Peterson
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines, La Jolla, CA, 92037, United States
| | - Linda M Bradley
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines, La Jolla, CA, 92037, United States
| | - Ze'ev A Ronai
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines, La Jolla, CA, 92037, United States.
| |
Collapse
|
479
|
Francescangeli F, De Angelis ML, Zeuner A. Dietary Factors in the Control of Gut Homeostasis, Intestinal Stem Cells, and Colorectal Cancer. Nutrients 2019; 11:nu11122936. [PMID: 31816977 PMCID: PMC6950549 DOI: 10.3390/nu11122936] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/21/2019] [Accepted: 11/27/2019] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is the third commonly diagnosed cancer and the second leading cause of cancer-related deaths worldwide. Global CRC burden is expected to increase by 60% in the next decade, with low-income countries experiencing an escalation of CRC incidence and mortality in parallel to the adoption of western lifestyles. CRC incidence is also sharply increasing in individuals younger than 50 years, often presenting at advanced stages and with aggressive features. Both genetic and environmental factors have been recognized as major contributors for the development of CRC, the latter including diet-related conditions such as chronic inflammation and obesity. In particular, a diet rich in fat and sugars (Western-style diet, WSD) has been shown to induce multiple pathophysiological changes in the intestine linked to an increased risk of CRC. In this scenario, dietary factors have been recently shown to play novel unexpected roles in the regulation of intestinal stem cells (ISCs) and of the gut microbiota, which represent the two main biological systems responsible for intestinal homeostasis. Furthermore, diet is increasingly recognized to play a key role in the neoplastic transformation of ISCs and in the metabolic regulation of colorectal cancer stem cells. This review illustrates novel discoveries on the role of dietary components in regulating intestinal homeostasis and colorectal tumorigenesis. Particular focus is dedicated to new areas of research with potential clinical relevance including the effect of food components on ISCs and cancer stem cells (CSCs), the existence of CRC-specific microbial signatures and the alterations of intestinal homeostasis potentially involved in early-onset CRC. New insights on the role of dietary factors in intestinal regulation will provide new tools not only for the prevention and early diagnosis of CRC but also for improving the effectiveness of current CRC therapies.
Collapse
|
480
|
Dąbrowska K, Abedon ST. Pharmacologically Aware Phage Therapy: Pharmacodynamic and Pharmacokinetic Obstacles to Phage Antibacterial Action in Animal and Human Bodies. Microbiol Mol Biol Rev 2019; 83:e00012-19. [PMID: 31666296 PMCID: PMC6822990 DOI: 10.1128/mmbr.00012-19] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The use of viruses infecting bacteria (bacteriophages or phages) to treat bacterial infections has been ongoing clinically for approximately 100 years. Despite that long history, the growing international crisis of resistance to standard antibiotics, abundant anecdotal evidence of efficacy, and one successful modern clinical trial of efficacy, this phage therapy is not yet a mainstream approach in medicine. One explanation for why phage therapy has not been subject to more widespread implementation is that phage therapy research, both preclinical and clinical, can be insufficiently pharmacologically aware. Consequently, here we consider the pharmacological obstacles to phage therapy effectiveness, with phages in phage therapy explicitly being considered to serve as drug equivalents. The study of pharmacology has traditionally been differentiated into pharmacokinetic and pharmacodynamic aspects. We therefore separately consider the difficulties that phages as virions can have in traveling through body compartments toward reaching their target bacteria (pharmacokinetics) and the difficulties that phages can have in exerting antibacterial activity once they have reached those bacteria (pharmacodynamics). The latter difficulties, at least in part, are functions of phage host range and bacterial resistance to phages. Given the apparently low toxicity of phages and the minimal side effects of phage therapy as practiced, phage therapy should be successful so long as phages can reach the targeted bacteria in sufficiently high numbers, adsorb, and then kill those bacteria. Greater awareness of what obstacles to this success generally or specifically can exist, as documented in this review, should aid in the further development of phage therapy toward wider use.
Collapse
Affiliation(s)
- Krystyna Dąbrowska
- Bacteriophage Laboratory, Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Stephen T Abedon
- Department of Microbiology, The Ohio State University, Mansfield, Ohio, USA
| |
Collapse
|
481
|
Vergara D, Simeone P, Damato M, Maffia M, Lanuti P, Trerotola M. The Cancer Microbiota: EMT and Inflammation as Shared Molecular Mechanisms Associated with Plasticity and Progression. JOURNAL OF ONCOLOGY 2019; 2019:1253727. [PMID: 31772577 PMCID: PMC6854237 DOI: 10.1155/2019/1253727] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/31/2019] [Indexed: 02/07/2023]
Abstract
With the advent of novel molecular platforms for high-throughput/next-generation sequencing, the communities of commensal and pathogenic microorganisms that inhabit the human body have been defined in depth. In the last decade, the role of microbiota-host interactions in driving human cancer plasticity and malignant progression has been well documented. Germ-free preclinical models provided an invaluable tool to demonstrate that the human microbiota can confer susceptibility to various types of cancer and can also modulate the host response to therapeutic treatments. Of interest, besides the detrimental effects of dysbiosis on cancer etiopathogenesis, specific microorganisms have been shown to exert protective activities against cancer growth. This has strong clinical implications, as restoration of the physiologic microbiota is being rapidly implemented as a novel anticancer therapeutic strategy. Here, we reviewed past and recent literature depicting the role of microbiota-host interactions in modulating key molecular mechanisms that drive human cancer plasticity and lead to malignant progression. We analyzed microbiota-host interactions occurring in the gut as well as in other anatomic sites, such as oral and nasal cavities, lungs, breast, esophagus, stomach, reproductive tract, and skin. We revealed a common ground of biological alterations and pathways modulated by a dysbiotic microbiota and potentially involved in the control of cancer progression. The molecular mechanisms most frequently affected by the pathogenic microorganisms to induce malignant progression involve epithelial-mesenchymal transition- (EMT-) dependent barrier alterations and tumor-promoting inflammation. This evidence may pave the way to better stratify high-risk cancer patients based on unique microenvironmental/microbial signatures and to develop novel, personalized, biological therapies.
Collapse
Affiliation(s)
- Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
- Laboratory of Clinical Proteomic, “Giovanni Paolo II” Hospital, ASL-Lecce, Italy
| | - Pasquale Simeone
- Department of Medicine and Aging Sciences, “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Laboratory of Cytomorphology, Center for Advanced Studies and Technology (CAST), “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Marina Damato
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
- Laboratory of Clinical Proteomic, “Giovanni Paolo II” Hospital, ASL-Lecce, Italy
| | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
- Laboratory of Clinical Proteomic, “Giovanni Paolo II” Hospital, ASL-Lecce, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Laboratory of Cytomorphology, Center for Advanced Studies and Technology (CAST), “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Marco Trerotola
- Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
482
|
Abolarinwa BA, Ibrahim RB, Huang YH. Conceptual Development of Immunotherapeutic Approaches to Gastrointestinal Cancer. Int J Mol Sci 2019; 20:E4624. [PMID: 31540435 PMCID: PMC6769557 DOI: 10.3390/ijms20184624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/14/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal (GI) cancer is one of the common causes of cancer-related death worldwide. Chemotherapy and/or immunotherapy are the current treatments, but some patients do not derive clinical benefits. Recently, studies from cancer molecular subtyping have revealed that tumor molecular biomarkers may predict the immunotherapeutic response of GI cancer patients. However, the therapeutic response of patients selected by the predictive biomarkers is suboptimal. The tumor immune-microenvironment apparently plays a key role in modulating these molecular-determinant predictive biomarkers. Therefore, an understanding of the development and recent advances in immunotherapeutic pharmacological intervention targeting tumor immune-microenvironments and their potential predictive biomarkers will be helpful to strengthen patient immunotherapeutic efficacy. The current review focuses on an understanding of how the host-microenvironment interactions and the predictive biomarkers can determine the efficacy of immune checkpoint inhibitors. The contribution of environmental pathogens and host immunity to GI cancer is summarized. A discussion regarding the clinical evidence of predictive biomarkers for clinical trial therapy design, current immunotherapeutic strategies, and the outcomes to GI cancer patients are highlighted. An understanding of the underlying mechanism can predict the immunotherapeutic efficacy and facilitate the future development of personalized therapeutic strategies targeting GI cancers.
Collapse
Affiliation(s)
- Bilikis Aderonke Abolarinwa
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Ridwan Babatunde Ibrahim
- Institute of Brain Science, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- Taiwan International Graduate Program (TIGP) in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei 11529, Taiwan.
| | - Yen-Hua Huang
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
- Comprehensive Cancer Center of Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
483
|
Cerf-Bensussan N. Microbiology and immunology: An ideal partnership for a tango at the gut surface-A tribute to Philippe Sansonetti. Cell Microbiol 2019; 21:e13097. [PMID: 31414516 PMCID: PMC7027583 DOI: 10.1111/cmi.13097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/02/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022]
Abstract
Over the past 20 years, the highly dynamic interactions that take place between hosts and the gut microbiota have emerged as a major determinant in health and disease. The complexity of the gut microbiota represents, however, a considerable challenge, and reductionist approaches are indispensable to define the contribution of individual bacteria to host responses and to dissect molecular mechanisms. In this tribute to Philippe Sansonetti, I would like to show how rewarding collaborations with microbiologists have guided our team of immunologists in the study of host–microbiota interactions and, thanks to the use of controlled colonisation experiments in gnotobiotic mice, toward the demonstration that segmented filamentous bacteria (SFB) are indispensable to drive the post‐natal maturation of the gut immune barrier in mice. The work led with Philippe Sansonetti to set up in vitro culture conditions has been one important milestone that laid the ground for in‐depth characterization of the molecular attributes of this unusual symbiont. Recent suggestions that SFB may be present in the human microbiota encourage further cross‐fertilising interactions between microbiologists and immunologists to define whether results from mice can be translated to humans and, if so, how SFB may be used to promote human intestinal defences against enteropathogens. Nurturing the competences to pursue this inspiring project is one legacy of Philippe Sansonetti.
Collapse
Affiliation(s)
- Nadine Cerf-Bensussan
- Laboratory of Intestinal Immunity, INSERM UMR 1163, Institut Imagine, Paris, France.,Université de Paris, Paris, France
| |
Collapse
|
484
|
Mosaddad SA, Tahmasebi E, Yazdanian A, Rezvani MB, Seifalian A, Yazdanian M, Tebyanian H. Oral microbial biofilms: an update. Eur J Clin Microbiol Infect Dis 2019; 38:2005-2019. [PMID: 31372904 DOI: 10.1007/s10096-019-03641-9] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/14/2019] [Indexed: 01/20/2023]
Abstract
Human oral cavity (mouth) hosts a complex microbiome consisting of bacteria, archaea, protozoa, fungi and viruses. These bacteria are responsible for two common diseases of the human mouth including periodontal (gum) and dental caries (tooth decay). Dental caries is caused by plaques, which are a community of microorganisms in biofilm format. Genetic and peripheral factors lead to variations in the oral microbiome. It has known that, in commensalism and coexistence between microorganisms and the host, homeostasis in the oral microbiome is preserved. Nonetheless, under some conditions, a parasitic relationship dominates the existing situation and the rise of cariogenic microorganisms results in dental caries. Utilizing advanced molecular biology techniques, new cariogenic microorganisms species have been discovered. The oral microbiome of each person is quite distinct. Consequently, commonly taken measures for disease prevention cannot be exactly the same for other individuals. The chance for developing tooth decay in individuals is dependent on factors such as immune system and oral microbiome which itself is affected by the environmental and genetic determinants. Early detection of dental caries, assessment of risk factors and designing personalized measure let dentists control the disease and obtain desired results. It is necessary for a dentist to consider dental caries as a result of a biological process to be targeted than treating the consequences of decay cavities. In this research, we critically review the literature and discuss the role of microbial biofilms in dental caries.
Collapse
Affiliation(s)
- Seyed Ali Mosaddad
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elahe Tahmasebi
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Yazdanian
- Department of Veterinary, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Alexander Seifalian
- Nanotechnology and Regenerative Medicine Commercialization Centre (Ltd), The London Bioscience Innovation Center, London, UK
| | - Mohsen Yazdanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Hamid Tebyanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
485
|
Peric A, Weiss J, Vulliemoz N, Baud D, Stojanov M. Bacterial Colonization of the Female Upper Genital Tract. Int J Mol Sci 2019; 20:E3405. [PMID: 31373310 PMCID: PMC6678922 DOI: 10.3390/ijms20143405] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/09/2019] [Accepted: 07/10/2019] [Indexed: 12/23/2022] Open
Abstract
Bacteria colonize most of the human body, and the female genital tract is not an exception. While the existence of a vaginal microbiota has been well established, the upper genital tract has been considered a sterile environment, with a general assumption that bacterial presence is associated with adverse clinical manifestation. However, recent metagenomic studies identified specific patterns of microbiota colonizing the uterus, fallopian tubes, ovaries, and placenta. These results need confirmation and further investigations since the data are only scarce. Bacterial colonization of these sites appears different from the vaginal one, despite evidence that vaginal bacteria could ascend to the upper genital tract through the cervix. Are these bacteria only commensal or do they play a role in the physiology of the female upper genital tract? Which are the genera that may have a negative and a positive impact on the female reproductive function? The aim of this review is to critically present all available data on upper genital tract microbiota and discuss its role in human reproduction, ranging from the technical aspects of these types of analyses to the description of specific bacterial genera. Although still very limited, research focusing on genital colonization of bacteria other than the vaginal milieu might bring novel insights into physiopathology of human reproduction.
Collapse
Affiliation(s)
- Adriana Peric
- Clinic for Reproductive Medicine and Gynecological Endocrinology, Cantonal Hospital Lucerne, 6000 Lucerne, Switzerland
| | - Jürgen Weiss
- Clinic for Reproductive Medicine and Gynecological Endocrinology, Cantonal Hospital Lucerne, 6000 Lucerne, Switzerland
| | - Nicolas Vulliemoz
- Fertility Medicine and Gynaecologic Endocrinology Unit, Department Woman-Mother-Child, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - David Baud
- Materno-fetal and Obstetrics Research Unit, Department Woman-Mother-Child, Lausanne University Hospital, 1011 Lausanne, Switzerland.
| | - Milos Stojanov
- Materno-fetal and Obstetrics Research Unit, Department Woman-Mother-Child, Lausanne University Hospital, 1011 Lausanne, Switzerland.
| |
Collapse
|
486
|
Godoy-Vitorino F. Human microbial ecology and the rising new medicine. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:342. [PMID: 31475212 DOI: 10.21037/atm.2019.06.56] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The first life forms on earth were Prokaryotic, and the evolution of all Eukaryotic life occurred with the help of bacteria. Animal-associated microbiota also includes members of the archaea, fungi, protists, and viruses. The genomes of this host-associated microbial life are called the microbiome. Across the mammalian tree, microbiomes guarantee the development of immunity, physiology, and resistance to pathogens. In humans, all surfaces and cavities are colonized by a microbiome, maintained by a careful balance between the host response and its colonizers-thus humans are considered now supraorganisms. These microbiomes supply essential ecosystem services that benefit health through homeostasis, and the loss of the indigenous microbiota leads to dysbiosis, which can have significant consequences to disease. This educational review aims to describe the importance of human microbial ecology, explain the ecological terms applied to the study of the human microbiome, developments within the cutting-edge microbiome field, and implications to diagnostic and treatment.
Collapse
Affiliation(s)
- Filipa Godoy-Vitorino
- Department of Microbiology and Medical Zoology, University of Puerto Rico School of Medicine, Medical Sciences Campus, San Juan, PR, USA
| |
Collapse
|
487
|
Morán-Ramos S, Villarreal-Molina MT, Canizales-Quinteros S. Host Genetics, Diet, and Microbiome: The Role of AMY1. Trends Microbiol 2019; 27:473-475. [PMID: 31056302 DOI: 10.1016/j.tim.2019.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 04/11/2019] [Indexed: 02/06/2023]
Abstract
Host gene variants selected by diet adaptation have been associated with the microbiome. Poole et al. (Cell Host Microbe 2019;25;553-564.E7) reported that AMY1 copy number, associated with obesity, also impacts microbiome composition and function. Complex genetics-diet-microbiome interactions and their effect on obesity could eventually translate into personalized nutrition.
Collapse
Affiliation(s)
- Sofía Morán-Ramos
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Mexico City 03940, Mexico; Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico
| | | | - Samuel Canizales-Quinteros
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico.
| |
Collapse
|
488
|
Freudenhammer M, Henneke P, Härtel C. Mikrobiom von Risikoneugeborenen und präventive Modifikation. Monatsschr Kinderheilkd 2019. [DOI: 10.1007/s00112-019-0684-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
489
|
Bassanini G, Ceccarani C, Borgo F, Severgnini M, Rovelli V, Morace G, Verduci E, Borghi E. Phenylketonuria Diet Promotes Shifts in Firmicutes Populations. Front Cell Infect Microbiol 2019; 9:101. [PMID: 31058098 PMCID: PMC6477998 DOI: 10.3389/fcimb.2019.00101] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/25/2019] [Indexed: 12/21/2022] Open
Abstract
Low-phenylalanine diet, the mainstay of treatment for phenylketonuria (PKU), has been shown to increase glycemic index and glycemic load, affecting the availability of substrates for microbial fermentation. Indeed, changes in the PKU gut microbiota compared with healthy controls have been previously reported. In this study we compared the gut microbial communities of children with PKU and with mild hyperphenylalaninemia (MHP, unrestricted diet). For each group, we enrolled 21 children (4-18 years old), for a total dataset of 42 subjects. We assessed dietary intake and performed gut microbiota analysis by sequencing the V3-V4 hypervariable regions of the 16S rRNA gene. Short chain fatty acids (SCFAs) were quantified by gas chromatographic analysis. While alpha-diversity analysis showed no significant differences between PKU and MHP groups, microbial community analysis highlighted a significant separation of the gut microbiota according to both unweighted (p = 0.008) and weighted Unifrac distances (p = 0.033). Major differences were seen within the Firmicutes phylum. Indeed, PKU children were depleted in Faecalibacterium spp. and enriched in Blautia spp. and Clostridium spp (family Lachnospiraceae). We found a divergent response of members of the Firmicutes phylum with respect to daily glycemic index, higher in PKU children. Faecalibacterium prausnitzii, unclassified Ruminococcaceae and, to a lesser extent Roseburia spp. negatively correlated with glycemic index, whereas unclassified Lachnospiraceae were positively associated. Indicator species analysis suggested F. prausnitzii be related to MHP status and Ruminococcus bromii to be associated with PKU. Despite PKU children having a higher vegetable and fiber intake, resembling a vegan diet, their gut microbial profile is different from the microbiota reported in the literature for individuals consuming a high-fiber/low-protein diet. Indeed, beneficial microorganisms, such as F. prausnitzii, considered a biomarker for a healthy status and one of the main butyrate producers, are depleted in PKU gut microbiota. We suggest that both the quality and quantity of carbohydrates ingested participate in determining the observed Firmicutes shifts on the PKU population.
Collapse
Affiliation(s)
- Giulia Bassanini
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Camilla Ceccarani
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Francesca Borgo
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Marco Severgnini
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Valentina Rovelli
- Department of Pediatrics, San Paolo Hospital, Università degli Studi di Milano, Milan, Italy
| | - Giulia Morace
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elvira Verduci
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
- Department of Pediatrics, San Paolo Hospital, Università degli Studi di Milano, Milan, Italy
| | - Elisa Borghi
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
490
|
Vandenplas Y, Savino F. Probiotics and Prebiotics in Pediatrics: What Is New? Nutrients 2019; 11:nu11020431. [PMID: 30791429 PMCID: PMC6412752 DOI: 10.3390/nu11020431] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/15/2019] [Indexed: 02/06/2023] Open
Affiliation(s)
- Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium.
| | - Francesco Savino
- Department of Pediatrics, Ospedale Infantile Regina Margherita, Azienda Ospedaliera, Universitaria Città della Salute e della Scienza di Torino, Piazza Polonia, 94, 10126 Turin, Italy.
| |
Collapse
|