751
|
Pham VT, Lacroix C, Braegger CP, Chassard C. Lactate-utilizing community is associated with gut microbiota dysbiosis in colicky infants. Sci Rep 2017; 7:11176. [PMID: 28894218 PMCID: PMC5593888 DOI: 10.1038/s41598-017-11509-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/25/2017] [Indexed: 12/12/2022] Open
Abstract
The aetiology of colic, a functional gastrointestinal disorder in infants, is not yet resolved. Different mechanisms have been suggested involving the gut microbiota and intermediate metabolites such as lactate. Lactate can be metabolized by lactate-utilizing bacteria (LUB) to form different end-products. Using a functional approach, we hypothesized that H2 production and accumulation by LUB is associated with the development of colic. The LUB communities in the feces of forty infants, including eight colicky infants, were characterized using a combination of culture- and molecular-based methods, and metabolite concentrations were measured by HPLC. Interactions among LUB strains isolated from feces were investigated with pure and mixed cultures using anaerobic techniques. We emphasized high prevalence of crying, flatulence, colic and positive correlations thereof in the first 3 months of life. Crying infants showed significantly higher ratio of LUB non-sulfate-reducing bacteria (LUB non-SRB) (H2-producer), to LUB SRB (H2-utilizer) at 3 months. Colicky infants had significantly higher number of H2-producing Eubacterium hallii at 2 weeks compared to non-colicky infants. We revealed the function of Desulfovibrio piger and Eubacterium limosum to reduce H2 accumulation in co-cultures with H2-producing Veillonella ratti. Our data suggest that the balance between H2-producing and H2-utilizing LUB might contribute to colic symptoms.
Collapse
Affiliation(s)
- Van T Pham
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland.,Division of Gastroenterology and Nutrition, University Children's Hospital Zurich, Zurich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland.
| | - Christian P Braegger
- Division of Gastroenterology and Nutrition, University Children's Hospital Zurich, Zurich, Switzerland
| | - Christophe Chassard
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland.,Université Clermont Auvergne, INRA, UMRF, F-1500, Aurillac, France
| |
Collapse
|
752
|
Thomson P, Medina DA, Garrido D. Human milk oligosaccharides and infant gut bifidobacteria: Molecular strategies for their utilization. Food Microbiol 2017; 75:37-46. [PMID: 30056961 DOI: 10.1016/j.fm.2017.09.001] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 08/30/2017] [Accepted: 09/02/2017] [Indexed: 02/08/2023]
Abstract
Breast milk is the gold standard in infant nutrition. In addition to provide essential nutrients for the newborn, it contains multiple bioactive molecules that provide protection and stimulate proper development. Human milk oligosaccharides (HMO) are complex carbohydrates abundant in breast milk. Intriguingly, these molecules do not provide energy to the infant. Instead, these oligosaccharides are key to guide and support the assembly of a healthy gut microbiome in the infant, dominated by beneficial gut microbes such as Bifidobacterium. New analytical methods for glycan analysis, and next-generation sequencing of microbial communities, have been instrumental in advancing our understanding of the positive role of breast milk oligosaccharides on the gut microbiome, and the genomics and molecular strategies of Bifidobacterium to utilize these oligosaccharides. Moreover, novel approaches to simulate the impact of HMO on the gut microbiome have been described and successfully validated, including the incorporation of synthetic HMO and bovine milk oligosaccharides to infant formula. This review discusses recent advances regarding the influence of HMO in promoting a healthy gut microbiome, with emphasis in the molecular basis of the enrichment in beneficial Bifidobacterium, and novel approaches to replicate the effect of HMO using synthetic or bovine oligosaccharides.
Collapse
Affiliation(s)
- Pamela Thomson
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago, Chile
| | - Daniel A Medina
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago, Chile
| | - Daniel Garrido
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago, Chile.
| |
Collapse
|
753
|
Franceschi C, Salvioli S, Garagnani P, de Eguileor M, Monti D, Capri M. Immunobiography and the Heterogeneity of Immune Responses in the Elderly: A Focus on Inflammaging and Trained Immunity. Front Immunol 2017; 8:982. [PMID: 28861086 PMCID: PMC5559470 DOI: 10.3389/fimmu.2017.00982] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/31/2017] [Indexed: 12/28/2022] Open
Abstract
Owing to its memory and plasticity, the immune system (IS) is capable of recording all the immunological experiences and stimuli it was exposed to. The combination of type, dose, intensity, and temporal sequence of antigenic stimuli that each individual is exposed to has been named “immunobiography.” This immunological history induces a lifelong continuous adaptation of the IS, which is responsible for the capability to mount strong, weak or no response to specific antigens, thus determining the large heterogeneity of immunological responses. In the last years, it is becoming clear that memory is not solely a feature of adaptive immunity, as it has been observed that also innate immune cells are provided with a sort of memory, dubbed “trained immunity.” In this review, we discuss the main characteristics of trained immunity as a possible contributor to inflammaging within the perspective of immunobiography, with particular attention to the phenotypic changes of the cell populations known to be involved in trained immunity. In conclusion, immunobiography emerges as a pervasive and comprehensive concept that could help in understanding and interpret the individual heterogeneity of immune responses (to infections and vaccinations) that becomes particularly evident at old age and could affect immunosenescence and inflammaging.
Collapse
Affiliation(s)
| | - Stefano Salvioli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Interdepartmental Centre 'L. Galvani' (CIG), University of Bologna, Bologna, Italy
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Interdepartmental Centre 'L. Galvani' (CIG), University of Bologna, Bologna, Italy
| | - Magda de Eguileor
- Department of Biotechnology and Life Science, University of Insubria, Varese, Italy
| | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Miriam Capri
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Interdepartmental Centre 'L. Galvani' (CIG), University of Bologna, Bologna, Italy
| |
Collapse
|
754
|
Torow N, Hornef MW. The Neonatal Window of Opportunity: Setting the Stage for Life-Long Host-Microbial Interaction and Immune Homeostasis. THE JOURNAL OF IMMUNOLOGY 2017; 198:557-563. [PMID: 28069750 DOI: 10.4049/jimmunol.1601253] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/17/2016] [Indexed: 02/06/2023]
Abstract
The existence of a neonatal window was first highlighted by epidemiological studies that revealed the particular importance of this early time in life for the susceptibility to immune-mediated diseases in humans. Recently, the first animal studies emerged that present examples of early-life exposure-triggered persisting immune events, allowing a detailed analysis of the factors that define this particular time period. The enteric microbiota and the innate and adaptive immune system represent prime candidates that impact on the pathogenesis of immune-mediated diseases and are known to reach a lasting homeostatic equilibrium following a dynamic priming period after birth. In this review, we outline the postnatal establishment of the microbiota and maturation of the innate and adaptive immune system and discuss examples of early-life exposure-triggered immune-mediated diseases that start to shed light on the critical importance of the early postnatal period for life-long immune homeostasis.
Collapse
Affiliation(s)
- Natalia Torow
- Institute of Medical Microbiology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| |
Collapse
|
755
|
Plunkett CH, Nagler CR. The Influence of the Microbiome on Allergic Sensitization to Food. THE JOURNAL OF IMMUNOLOGY 2017; 198:581-589. [PMID: 28069753 DOI: 10.4049/jimmunol.1601266] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 08/23/2016] [Indexed: 02/06/2023]
Abstract
The alarming increase in the incidence and severity of food allergies has coincided with lifestyle changes in Western societies, such as dietary modifications and increased antibiotic use. These demographic shifts have profoundly altered the coevolved relationship between host and microbiota, depleting bacterial populations critical for the maintenance of mucosal homeostasis. There is increasing evidence that the dysbiosis associated with sensitization to food fails to stimulate protective tolerogenic pathways, leading to the development of the type 2 immune responses that characterize allergic disease. Defining the role of beneficial allergy-protective members of the microbiota in the regulation of tolerance to food has exciting potential for new interventions to treat dietary allergies by modulation of the microbiota.
Collapse
Affiliation(s)
| | - Cathryn R Nagler
- Department of Pathology, The University of Chicago, Chicago, IL 60637; and .,Committee on Immunology, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
756
|
Nogacka A, Salazar N, Suárez M, Milani C, Arboleya S, Solís G, Fernández N, Alaez L, Hernández-Barranco AM, de Los Reyes-Gavilán CG, Ventura M, Gueimonde M. Impact of intrapartum antimicrobial prophylaxis upon the intestinal microbiota and the prevalence of antibiotic resistance genes in vaginally delivered full-term neonates. MICROBIOME 2017; 5:93. [PMID: 28789705 PMCID: PMC5549288 DOI: 10.1186/s40168-017-0313-3] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 07/21/2017] [Indexed: 05/14/2023]
Abstract
BACKGROUND Disturbances in the early establishment of the intestinal microbiota may produce important implications for the infant's health and for the risk of disease later on. Different perinatal conditions may be affecting the development of the gut microbiota. Some of them, such as delivery mode or feeding habits, have been extensively assessed whereas others remain to be studied, being critical to identify their impact on the microbiota and, if any, to minimize it. Antibiotics are among the drugs most frequently used in early life, the use of intrapartum antimicrobial prophylaxis (IAP), present in over 30% of deliveries, being the most frequent source of exposure. However, our knowledge on the effects of IAP on the microbiota establishment is still limited. The aim of the present work was to evaluate the impact of IAP investigating a cohort of 40 full-term vaginally delivered infants born after an uncomplicated pregnancy, 18 of which were born from mothers receiving IAP. RESULTS Fecal samples were collected at 2, 10, 30, and 90 days of age. We analyzed the composition of the fecal microbiota during the first 3 months of life by 16S rRNA gene sequencing and quantified fecal short chain fatty acids by gas chromatography. The presence of genes for resistance to antibiotics was determined by PCR in the samples from 1-month-old infants. Our results showed an altered pattern of intestinal microbiota establishment in IAP infants during the first weeks of life, with lower relative proportions of Actinobacteria and Bacteroidetes and increased of Preoteobacteria and Firmicutes. A delay in the increase on the levels of acetate was observed in IAP infants. The analyses of specific antibiotic resistance genes showed a higher occurrence of some β-lactamase coding genes in infants whose mothers received IAP. CONCLUSIONS Our results indicate an effect of IAP on the establishing early microbiota during the first months of life, which represent a key moment for the development of the microbiota-induced host homeostasis. Understanding the impact of IAP in the gut microbiota development is essential for developing treatments to minimize it, favoring a proper gut microbiota development in IAP-exposed neonates.
Collapse
Affiliation(s)
- Alicja Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias. Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Ctra. Infiesto s/n, 33300, Villaviciosa, Asturias, Spain
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias. Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Ctra. Infiesto s/n, 33300, Villaviciosa, Asturias, Spain
| | - Marta Suárez
- Pediatrics Service, Hospital Universitario Central de Asturias, SESPA, Oviedo, Asturias, Spain
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma, Parma, Italy
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias. Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Ctra. Infiesto s/n, 33300, Villaviciosa, Asturias, Spain
- Current address: APC Microbiome Institute, University College Cork. Cork, Ireland & Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Gonzalo Solís
- Pediatrics Service, Hospital Universitario Central de Asturias, SESPA, Oviedo, Asturias, Spain
| | - Nuria Fernández
- Pediatrics Service, Hospital Universitario Central de Asturias, SESPA, Oviedo, Asturias, Spain
| | - Lidia Alaez
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias. Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Ctra. Infiesto s/n, 33300, Villaviciosa, Asturias, Spain
| | - Ana M Hernández-Barranco
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias. Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Ctra. Infiesto s/n, 33300, Villaviciosa, Asturias, Spain
| | - Clara G de Los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias. Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Ctra. Infiesto s/n, 33300, Villaviciosa, Asturias, Spain
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma, Parma, Italy
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias. Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Ctra. Infiesto s/n, 33300, Villaviciosa, Asturias, Spain.
| |
Collapse
|
757
|
Walker RW, Clemente JC, Peter I, Loos RJF. The prenatal gut microbiome: are we colonized with bacteria in utero? Pediatr Obes 2017; 12 Suppl 1:3-17. [PMID: 28447406 PMCID: PMC5583026 DOI: 10.1111/ijpo.12217] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/17/2017] [Accepted: 02/02/2017] [Indexed: 12/11/2022]
Abstract
The colonization of the gut with microbes in early life is critical to the developing newborn immune system, metabolic function and potentially future health. Maternal microbes are transmitted to offspring during childbirth, representing a key step in the colonization of the infant gut. Studies of infant meconium suggest that bacteria are present in the foetal gut prior to birth, meaning that colonization could occur prenatally. Animal studies have shown that prenatal transmission of microbes to the foetus is possible, and physiological changes observed in pregnant mothers indicate that in utero transfer is likely in humans as well. However, direct evidence of in utero transfer of bacteria in humans is lacking. Understanding the timing and mechanisms involved in the first colonization of the human gut is critical to a comprehensive understanding of the early life gut microbiome. This review will discuss the evidence supporting in utero transmission of microbes from mother to infants. We also review sources of transferred bacteria, physiological mechanisms of transfer and modifiers of maternal microbiomes and their potential role in early life infant health. Well-designed longitudinal birth studies that account for established modifiers of the gut microbiome are challenging, but will be necessary to confirm in utero transfer and further our knowledge of the prenatal microbiome.
Collapse
Affiliation(s)
- Ryan W Walker
- Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10025, USA
| | - Jose C Clemente
- Genetics and Genomic Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY 10025, USA
| | - Inga Peter
- Genetics and Genomic Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY 10025, USA
| | - Ruth JF Loos
- Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10025, USA
| |
Collapse
|
758
|
Nagpal R, Tsuji H, Takahashi T, Nomoto K, Kawashima K, Nagata S, Yamashiro Y. Ontogenesis of the Gut Microbiota Composition in Healthy, Full-Term, Vaginally Born and Breast-Fed Infants over the First 3 Years of Life: A Quantitative Bird's-Eye View. Front Microbiol 2017; 8:1388. [PMID: 28785253 PMCID: PMC5519616 DOI: 10.3389/fmicb.2017.01388] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/10/2017] [Indexed: 12/26/2022] Open
Abstract
Early-life intestinal microbiota development is crucial for host’s long-term health and is influenced by many factors including gestational age, birth and feeding modes, birth environment, ethnic/geographical background, etc. However, ‘quantitative’ data on the actual population levels of gut bacterial communities when these influences are controlled for is relatively rare. Herein, we demonstrate a quantitative perspective of microbiota development in natural and healthy milieus, i.e., in healthy, full-term, vaginally born and breast-fed infants (n = 19) born at same clinic. Fecal microbiota at age 1 and 7 days, 1, 3, and 6 months and 3 years is quantified using highly sensitive reverse-transcription-quantitative-PCR assays targeting bacterial rRNA molecules. At day 1, we detect one or more bacteria in all (100%) of the babies, wherein the microbiota is composed mainly of enterobacteria (35%), Bacteroides fragilis group (23%), enterococci (18%), staphylococci (13%), and bifidobacteria (9%). Altogether, facultative anaerobes predominate during first few weeks whereafter obligate anaerobes including bifidobacteria, B. fragilis group, Clostridium coccoides group, and Clostridium leptum subgroup gradually start prevailing. At 3 years, the composition is represented almost entirely (99%) by obligate anaerobes including C. leptum subgroup (34%), bifidobacteria (22%), B. fragilis group (21%), C. coccoides group (17%), Atopobium cluster (4%), and Prevotella (1%). The overall obligate/facultative proportion is 32/68, 37/63, 54/46, 70/30, 64/36, and 99/1% at 1 and 7 days, 1, 3, and 6 months and 3 years, respectively. However, interestingly, considerable individual-specific variations in the obligate/facultative ratios as well as in the proportions of Firmicutes, Bacteroides, Actinobacteria, and Proteobacteria communities are seen among these babies. This disparity even within this highly homogenous cohort manifests the magnitude of diverse patterns of gut microbiota configuration and hence underpins the importance of considering not only the gestational age, birth, and feeding modes, and ethnic/geographical background but also other potential outstanding factors when investigating the elements shaping the early microbiota development. In summary, the data demonstrate a quantitative bird’s-eye view of the ontogenesis of early-life gut microbiota in typically natural and healthy milieus and should be informative and facilitative for future studies exploring various aspects of the human gut microbiota.
Collapse
Affiliation(s)
- Ravinder Nagpal
- Probiotics Research Laboratory, Juntendo University Graduate School of MedicineTokyo, Japan
| | - Hirokazu Tsuji
- Probiotics Research Laboratory, Juntendo University Graduate School of MedicineTokyo, Japan.,Yakult Central InstituteKunitachi, Japan
| | - Takuya Takahashi
- Probiotics Research Laboratory, Juntendo University Graduate School of MedicineTokyo, Japan.,Yakult Central InstituteKunitachi, Japan
| | - Koji Nomoto
- Probiotics Research Laboratory, Juntendo University Graduate School of MedicineTokyo, Japan.,Yakult Central InstituteKunitachi, Japan
| | | | - Satoru Nagata
- Department of Pediatrics, School of Medicine, Tokyo Women's Medical UniversityShinjuku, Japan
| | - Yuichiro Yamashiro
- Probiotics Research Laboratory, Juntendo University Graduate School of MedicineTokyo, Japan
| |
Collapse
|
759
|
Mima K, Ogino S, Nakagawa S, Sawayama H, Kinoshita K, Krashima R, Ishimoto T, Imai K, Iwatsuki M, Hashimoto D, Baba Y, Sakamoto Y, Yamashita YI, Yoshida N, Chikamoto A, Ishiko T, Baba H. The role of intestinal bacteria in the development and progression of gastrointestinal tract neoplasms. Surg Oncol 2017; 26:368-376. [PMID: 29113654 DOI: 10.1016/j.suronc.2017.07.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 07/09/2017] [Accepted: 07/20/2017] [Indexed: 12/12/2022]
Abstract
More than 100 trillion microorganisms inhabit the human intestinal tract and play important roles in health conditions and diseases, including cancer. Accumulating evidence demonstrates that specific bacteria and bacterial dysbiosis in the gastrointestinal tract can potentiate the development and progression of gastrointestinal tract neoplasms by damaging DNA, activating oncogenic signaling pathways, producing tumor-promoting metabolites such as secondary bile acids, and suppressing antitumor immunity. Other bacterial species have been shown to produce short-chain fatty acids such as butyrate, which can suppress inflammation and carcinogenesis in the gastrointestinal tract. Consistent with these lines of evidence, clinical studies using metagenomic analyses have shown associations of specific bacteria and bacterial dysbiosis with gastrointestinal tract cancers, including esophageal, gastric, and colorectal cancers. Emerging data demonstrate that intestinal bacteria can modulate the efficacy of cancer chemotherapies and novel targeted immunotherapies such as anti-CTLA4 and anti-CD274 therapies, the process of absorption, and the occurrence of complications after gastrointestinal surgery. A better understanding of the mechanisms by which the gut microbiota influence tumor development and progression in the intestine would provide opportunities to develop new prevention and treatment strategies for patients with gastrointestinal tract cancers by targeting the intestinal microflora.
Collapse
Affiliation(s)
- Kosuke Mima
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Shuji Ogino
- Division of MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shigeki Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Sawayama
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Koichi Kinoshita
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Ryuichi Krashima
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Katsunori Imai
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Daisuke Hashimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yasuo Sakamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yo-Ichi Yamashita
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Akira Chikamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Takatoshi Ishiko
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
760
|
Xiong W, Brown CT, Morowitz MJ, Banfield JF, Hettich RL. Genome-resolved metaproteomic characterization of preterm infant gut microbiota development reveals species-specific metabolic shifts and variabilities during early life. MICROBIOME 2017; 5:72. [PMID: 28693612 PMCID: PMC5504695 DOI: 10.1186/s40168-017-0290-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/19/2017] [Indexed: 05/16/2023]
Abstract
BACKGROUND Establishment of the human gut microbiota begins at birth. This early-life microbiota development can impact host physiology during infancy and even across an entire life span. However, the functional stability and population structure of the gut microbiota during initial colonization remain poorly understood. Metaproteomics is an emerging technology for the large-scale characterization of metabolic functions in complex microbial communities (gut microbiota). RESULTS We applied a metagenome-informed metaproteomic approach to study the temporal and inter-individual differences of metabolic functions during microbial colonization of preterm human infants' gut. By analyzing 30 individual fecal samples, we identified up to 12,568 protein groups for each of four infants, including both human and microbial proteins. With genome-resolved matched metagenomics, proteins were confidently identified at the species/strain level. The maximum percentage of the proteome detected for the abundant organisms was ~45%. A time-dependent increase in the relative abundance of microbial versus human proteins suggested increasing microbial colonization during the first few weeks of early life. We observed remarkable variations and temporal shifts in the relative protein abundances of each organism in these preterm gut communities. Given the dissimilarity of the communities, only 81 microbial EggNOG orthologous groups and 57 human proteins were observed across all samples. These conserved microbial proteins were involved in carbohydrate, energy, amino acid and nucleotide metabolism while conserved human proteins were related to immune response and mucosal maturation. We identified seven proteome clusters for the communities and showed infant gut proteome profiles were unstable across time and not individual-specific. Applying a gut-specific metabolic module (GMM) analysis, we found that gut communities varied primarily in the contribution of nutrient (carbohydrates, lipids, and amino acids) utilization and short-chain fatty acid production. CONCLUSIONS Overall, this study reports species-specific proteome profiles and metabolic functions of human gut microbiota during early colonization. In particular, our work contributes to reveal microbiota-associated shifts and variations in the metabolism of three major nutrient sources and short-chain fatty acid during colonization of preterm infant gut.
Collapse
Affiliation(s)
- Weili Xiong
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN USA
| | - Christopher T. Brown
- Department of Earth and Planetary Science, University of California, Berkeley, CA USA
| | | | - Jillian F. Banfield
- Department of Earth and Planetary Science, University of California, Berkeley, CA USA
| | - Robert L. Hettich
- Chemical Sciences Division, Oak Ridge National Laboratory, Bethel Valley Rd, Oak Ridge, TN 37831 USA
| |
Collapse
|
761
|
Han YY, Forno E, Badellino HA, Celedón JC. Antibiotic Use in Early Life, Rural Residence, and Allergic Diseases in Argentinean Children. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2017; 5:1112-1118.e2. [PMID: 28174014 PMCID: PMC5503768 DOI: 10.1016/j.jaip.2016.12.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/13/2016] [Accepted: 12/28/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND Little is known about differential effects of antibiotic use on allergic diseases in rural versus urban environments. OBJECTIVE To examine whether area of residence in the first year of life modifies the relation between antibiotic use in early life and allergic diseases during childhood. METHODS Cross-sectional study of allergic diseases in 1517 children (ages 6-7 years) attending 101 schools in urban and rural areas of San Francisco (Córdoba, Argentina). Current asthma, wheeze, and allergic rhinoconjunctivitis were defined on the basis of responses to a validated questionnaire from the International Study of Asthma and Allergies in Childhood. Multivariate logistic regression was used for the analysis of antibiotic use and allergic diseases. RESULTS After adjustment for paracetamol use, bronchiolitis, and other covariates, antibiotic use in the first year of life was associated with increased odds of current wheeze (odds ratio [OR], 1.8; 95% CI, 1.3-2.6) and allergic rhinoconjunctivitis (OR, 1.9; 95% CI, 1.3-2.7). After stratification by area of residence, antibiotic use was associated with current wheeze (OR, 2.4; 95% CI, 1.5-4.0) and allergic rhinoconjunctivitis (OR, 2.1; 95% CI, 1.3-3.4) among children who lived in an urban area in their first year of life, but not among those who lived in a rural area in their first year of life. CONCLUSIONS Early-life antibiotic use is associated with current wheeze and allergic rhinoconjunctivitis in Argentinean children who lived in urban areas during their first year of life. Exposure to a rural environment early in life may protect against the adverse effects of antibiotics on atopic diseases in children.
Collapse
Affiliation(s)
- Yueh-Ying Han
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pa
| | - Erick Forno
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pa
| | - Héctor A Badellino
- Department of Pediatric Respiratory Medicine, Regional Eastern Clinic, San Francisco, Córdoba, Argentina
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pa.
| |
Collapse
|
762
|
Lai JCY, Rocha-Ferreira E, Ek CJ, Wang X, Hagberg H, Mallard C. Immune responses in perinatal brain injury. Brain Behav Immun 2017; 63:210-223. [PMID: 27865947 DOI: 10.1016/j.bbi.2016.10.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/28/2016] [Accepted: 10/30/2016] [Indexed: 12/13/2022] Open
Abstract
The perinatal period has often been described as immune deficient. However, it has become clear that immune responses in the neonate following exposure to microbes or as a result of tissue injury may be substantial and play a role in perinatal brain injury. In this article we will review the immune cell composition under normal physiological conditions in the perinatal period, both in the human and rodent. We will summarize evidence of the inflammatory responses to stimuli and discuss how neonatal immune activation, both in the central nervous system and in the periphery, may contribute to perinatal hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Jacqueline C Y Lai
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Eridan Rocha-Ferreira
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - C Joakim Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Xiaoyang Wang
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Henrik Hagberg
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden.
| |
Collapse
|
763
|
Becerra-Díaz M, Wills-Karp M, Heller NM. New perspectives on the regulation of type II inflammation in asthma. F1000Res 2017; 6:1014. [PMID: 28721208 PMCID: PMC5497827 DOI: 10.12688/f1000research.11198.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/16/2017] [Indexed: 12/12/2022] Open
Abstract
Asthma is a chronic inflammatory disease of the lungs which has been thought to arise as a result of inappropriately directed T helper type-2 (Th2) immune responses of the lungs to otherwise innocuous inhaled antigens. Current asthma therapeutics are directed towards the amelioration of downstream consequences of type-2 immune responses (i.e. β-agonists) or broad-spectrum immunosuppression (i.e. corticosteroids). However, few approaches to date have been focused on the primary prevention of immune deviation. Advances in molecular phenotyping reveal heterogeneity within the asthmatic population with multiple endotypes whose varying expression depends on the interplay between numerous environmental factors and the inheritance of a broad range of susceptibility genes. The most common endotype is one described as "type-2-high" (i.e. high levels of interleukin [IL]-13, eosinophilia, and periostin). The identification of multiple endotypes has provided a potential explanation for the observations that therapies directed at typical Th2 cytokines (IL-4, IL-5, and IL-13) and their receptors have often fallen short when they were tested in a diverse group of asthmatic patients without first stratifying based on disease endotype or severity. However, despite the incorporation of endotype-dependent stratification schemes into clinical trial designs, variation in drug responses are still apparent, suggesting that additional genetic/environmental factors may be contributing to the diversity in drug efficacy. Herein, we will review recent advances in our understanding of the complex pathways involved in the initiation and regulation of type-2-mediated immune responses and their modulation by host factors (genetics, metabolic status, and the microbiome). Particular consideration will be given to how this knowledge could pave the way for further refinement of disease endotypes and/or the development of novel therapeutic strategies for the treatment of asthma .
Collapse
Affiliation(s)
- Mireya Becerra-Díaz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Marsha Wills-Karp
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Nicola M. Heller
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
764
|
Aitoro R, Paparo L, Amoroso A, Di Costanzo M, Cosenza L, Granata V, Di Scala C, Nocerino R, Trinchese G, Montella M, Ercolini D, Berni Canani R. Gut Microbiota as a Target for Preventive and Therapeutic Intervention against Food Allergy. Nutrients 2017; 9:nu9070672. [PMID: 28657607 PMCID: PMC5537787 DOI: 10.3390/nu9070672] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/15/2017] [Accepted: 06/23/2017] [Indexed: 12/24/2022] Open
Abstract
The gut microbiota plays a pivotal role in immune system development and function. Modification in the gut microbiota composition (dysbiosis) early in life is a critical factor affecting the development of food allergy. Many environmental factors including caesarean delivery, lack of breast milk, drugs, antiseptic agents, and a low-fiber/high-fat diet can induce gut microbiota dysbiosis, and have been associated with the occurrence of food allergy. New technologies and experimental tools have provided information regarding the importance of select bacteria on immune tolerance mechanisms. Short-chain fatty acids are crucial metabolic products of gut microbiota responsible for many protective effects against food allergy. These compounds are involved in epigenetic regulation of the immune system. These evidences provide a foundation for developing innovative strategies to prevent and treat food allergy. Here, we present an overview on the potential role of gut microbiota as the target of intervention against food allergy.
Collapse
Affiliation(s)
- Rosita Aitoro
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Lorella Paparo
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Antonio Amoroso
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Margherita Di Costanzo
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Linda Cosenza
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Viviana Granata
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Carmen Di Scala
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Rita Nocerino
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Giovanna Trinchese
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Mariangela Montella
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Danilo Ercolini
- Department of Agricultural Sciences, Division of Microbiology, University of Naples "Federico II", 80055 Portici, Italy.
- Task Force on Microbiome Studies, University of Naples "Federico II", 80131 Naples, Italy.
| | - Roberto Berni Canani
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II", 80131 Naples, Italy.
- Task Force on Microbiome Studies, University of Naples "Federico II", 80131 Naples, Italy.
- European Laboratory for the Investigation of Food Induced Diseases, University of Naples "Federico II", 80131 Naples, Italy.
- CEINGE Advanced Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy.
| |
Collapse
|
765
|
Wen L, Duffy A. Factors Influencing the Gut Microbiota, Inflammation, and Type 2 Diabetes. J Nutr 2017; 147:1468S-1475S. [PMID: 28615382 DOI: 10.3945/jn.116.240754] [Citation(s) in RCA: 248] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/12/2016] [Accepted: 01/11/2017] [Indexed: 12/17/2022] Open
Abstract
The gut microbiota is a complex community of bacteria residing in the intestine. Animal models have demonstrated that several factors contribute to and can significantly alter the composition of the gut microbiota, including genetics; the mode of delivery at birth; the method of infant feeding; the use of medications, especially antibiotics; and the diet. There may exist a gut microbiota signature that promotes intestinal inflammation and subsequent systemic low-grade inflammation, which in turn promotes the development of type 2 diabetes. There are preliminary studies that suggest that the consumption of probiotic bacteria such as those found in yogurt and other fermented milk products can beneficially alter the composition of the gut microbiome, which in turn changes the host metabolism. Obesity, insulin resistance, fatty liver disease, and low-grade peripheral inflammation are more prevalent in patients with low α diversity in the gut microbiome than they are in patients with high α diversity. Fermented milk products, such as yogurt, deliver a large number of lactic acid bacteria to the gastrointestinal tract. They may modify the intestinal environment, including inhibiting lipopolysaccharide production and increasing the tight junctions of gut epithelia cells.
Collapse
Affiliation(s)
- Li Wen
- Section of Endocrinology and
| | - Andrew Duffy
- Department of Surgery, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
766
|
Miyamoto M, Gouvêa AFTB, Ono E, Succi RCM, Pahwa S, Moraes-Pinto MID. Immune development in HIV-exposed uninfected children born to HIV-infected women. Rev Inst Med Trop Sao Paulo 2017; 59:e30. [PMID: 28591258 PMCID: PMC5459537 DOI: 10.1590/s1678-9946201759030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 02/22/2017] [Indexed: 01/15/2023] Open
Abstract
Immunological and clinical findings suggestive of some immune dysfunction have been reported among HIV-exposed uninfected (HEU) children and adolescents. Whether these defects are persistent or transitory is still unknown. HEU pediatric population at birth, 12 months, 6-12 years were evaluated in comparison to healthy age-matched HIV-unexposed controls. Plasma levels of LPS, sCD14, cytokines, lymphocyte immunophenotyping and T-cell receptor excision circles (TREC) were assessed. HEU and controls had similar LPS levels, which remained low from birth to 6-12 years; for plasma sCD14, IL-2, IL-6, IL-7, IL-10, IL-12p70, IL-13, IL-17, IFN-γ, TNF-α, G-CSF, GM-CSF and MCP-1, which increased from birth to 12 months and then decreased at 6-12 years; and for TREC/106 PBMC at birth in HEU and controls. By contrast, plasma MIP-1β levels were lower in HEU than in controls (p=0.009) at 12 months, and IL-4 levels were higher in HEU than controls (p=0.04) at 6-12 years. Immune activation was higher in HEU at 12 months and at 6-12 years than controls based on frequencies of CD38+HLA-DR+CD8+T cells (p=0.05) and of CD38+HLA-DR+CD4+T cells (p=0.006). Resting memory and activated mature B cells increased from birth to 6-12 years in both groups. The development of the immune system in vertically HEU individuals is comparable to the general population in most parameters, but subtle or transient differences exist. Their role in influencing clinical incidences in HEU is unknown.
Collapse
Affiliation(s)
- Maristela Miyamoto
- Universidade Federal de São Paulo, Departamento de Pediatria, São Paulo, São Paulo, Brazil
| | - Aída F T B Gouvêa
- Universidade Federal de São Paulo, Departamento de Pediatria, São Paulo, São Paulo, Brazil
| | - Erika Ono
- Universidade Federal de São Paulo, Departamento de Pediatria, São Paulo, São Paulo, Brazil
| | - Regina Célia M Succi
- Universidade Federal de São Paulo, Departamento de Pediatria, São Paulo, São Paulo, Brazil
| | - Savita Pahwa
- University of Miami, Department of Medicine, Miami, USA
| | | |
Collapse
|
767
|
Eaimworawuthikul S, Thiennimitr P, Chattipakorn N, Chattipakorn SC. Diet-induced obesity, gut microbiota and bone, including alveolar bone loss. Arch Oral Biol 2017; 78:65-81. [DOI: 10.1016/j.archoralbio.2017.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/09/2017] [Accepted: 02/06/2017] [Indexed: 02/07/2023]
|
768
|
Microbiota in T-cell homeostasis and inflammatory diseases. Exp Mol Med 2017; 49:e340. [PMID: 28546563 PMCID: PMC5454441 DOI: 10.1038/emm.2017.36] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 01/02/2017] [Indexed: 12/11/2022] Open
Abstract
The etiology of disease pathogenesis can be largely explained by genetic variations and several types of environmental factors. In genetically disease-susceptible individuals, subsequent environmental triggers may induce disease development. The human body is colonized by complex commensal microbes that have co-evolved with the host immune system. With the adaptation to modern lifestyles, its composition has changed depending on host genetics, changes in diet, overuse of antibiotics against infection and elimination of natural enemies through the strengthening of sanitation. In particular, commensal microbiota is necessary in the development, induction and function of T cells to maintain host immune homeostasis. Alterations in the compositional diversity and abundance levels of microbiota, known as dysbiosis, can trigger several types of autoimmune and inflammatory diseases through the imbalance of T-cell subpopulations, such as Th1, Th2, Th17 and Treg cells. Recently, emerging evidence has identified that dysbiosis is involved in the progression of rheumatoid arthritis, type 1 and 2 diabetic mellitus, and asthma, together with dysregulated T-cell subpopulations. In this review, we will focus on understanding the complicated microbiota-T-cell axis between homeostatic and pathogenic conditions and elucidate important insights for the development of novel targets for disease therapy.
Collapse
|
769
|
Amenyogbe N, Kollmann TR, Ben-Othman R. Early-Life Host-Microbiome Interphase: The Key Frontier for Immune Development. Front Pediatr 2017; 5:111. [PMID: 28596951 PMCID: PMC5442244 DOI: 10.3389/fped.2017.00111] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/28/2017] [Indexed: 12/14/2022] Open
Abstract
Human existence can be viewed as an "animal in a microbial world." A healthy interaction of the human host with the microbes in and around us heavily relies on a well-functioning immune system. As development of both the microbiota and the host immune system undergo rapid changes in early life, it is not surprising that even minor alterations during this co-development can have profound consequences. Scrutiny of existing data regarding pre-, peri-, as well as early postnatal modulators of newborn microbiota indeed suggest strong associations with several immune-mediated diseases with onset far beyond the newborn period. We here summarize these data and extract overarching themes. This same effort in turn sets the stage to guide effective countermeasures, such as probiotic administration. The objective of our review is to highlight the interaction of host immune ontogeny with the developing microbiome in early life as a critical window of susceptibility for lifelong disease, as well as to identify the enormous potential to protect and promote lifelong health by specifically targeting this window of opportunity.
Collapse
Affiliation(s)
- Nelly Amenyogbe
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tobias R. Kollmann
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Rym Ben-Othman
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
770
|
Mima K, Nakagawa S, Sawayama H, Ishimoto T, Imai K, Iwatsuki M, Hashimoto D, Baba Y, Yamashita YI, Yoshida N, Chikamoto A, Baba H. The microbiome and hepatobiliary-pancreatic cancers. Cancer Lett 2017; 402:9-15. [PMID: 28527946 DOI: 10.1016/j.canlet.2017.05.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 04/29/2017] [Accepted: 05/11/2017] [Indexed: 02/07/2023]
Abstract
The human intestinal microbiome encompasses at least 100 trillion microorganisms that can influence host immunity and disease conditions, including cancer. Hepatobiliary and pancreatic cancers have been associated with poor prognosis owing to their high level of tumor invasiveness, distant metastasis, and resistance to conventional treatment options, such as chemotherapy. Accumulating evidence from animal models suggests that specific microbes and microbial dysbiosis can potentiate hepatobiliary-pancreatic tumor development by damaging DNA, activating oncogenic signaling pathways, and producing tumor-promoting metabolites. Emerging evidence suggests that the gut microbiota may influence not only the efficacy of cancer chemotherapies and novel targeted immunotherapies such as anti-CTLA4 and anti-CD274 therapies but also the occurrence of postoperative complications after hepatobiliary and pancreatic surgery, which have been associated with tumor recurrence and worse patient survival in hepatobiliary-pancreatic cancers. Hence, a better understanding of roles of the gut microbiota in the development and progression of hepatobiliary-pancreatic tumors may open opportunities to develop new prevention and treatment strategies for patients with hepatobiliary-pancreatic cancer through manipulating the gut microbiota by diet, lifestyle, antibiotics, and pro- and prebiotics.
Collapse
Affiliation(s)
- Kosuke Mima
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Shigeki Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Sawayama
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Katsunori Imai
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Daisuke Hashimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yo-Ichi Yamashita
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Akira Chikamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
771
|
Tamburini S, Clemente JC. Gut microbiota: Neonatal gut microbiota induces lung immunity against pneumonia. Nat Rev Gastroenterol Hepatol 2017; 14:263-264. [PMID: 28325929 DOI: 10.1038/nrgastro.2017.34] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Sabrina Tamburini
- Icahn Institute for Genomics &Multiscale Biology, Department of Genetics &Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, New York 10029, USA
| | - Jose C Clemente
- Icahn Institute for Genomics &Multiscale Biology, Department of Genetics &Genomic Sciences, and the Precision Immunology Institute and Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, New York 10029, USA
| |
Collapse
|
772
|
Abstract
The intestinal microbiota consists of a dynamic organization of bacteria, viruses, archaea, and fungal species essential for maintaining gut homeostasis and protecting the host against pathogenic invasion. When dysregulated, the intestinal microbiota can contribute to colorectal cancer development. Though the microbiota is multifaceted in its ability to induce colorectal cancer, this review will focus on the capability of the microbiota to induce colorectal cancer through the modulation of immune function and the production of microbial-derived metabolites. We will also explore an experimental technique that is revolutionizing intestinal research. By elucidating the interactions of microbial species with epithelial tissue, and allowing for drug screening of patients with colorectal cancers, organoid development is a novel culturing technique that is innovating intestinal research. As a cancer that remains one of the leading causes of cancer-related deaths worldwide, it is imperative that scientific findings are translated into the creation of effective therapeutics to treat colorectal cancer.
Collapse
Affiliation(s)
- Sofia Oke
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Alberto Martin
- Department of Immunology, University of Toronto, 1 King’s College Cir, MSB 7302, Toronto, Ontario M5S 1A1, Canada
| |
Collapse
|
773
|
Affiliation(s)
- M Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - J Penders
- Department of Medical Microbiology, NUTRIM School of Nutrition and Translational Research in Metabolism & Caphri School for Public Health and Primary Care, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
774
|
Li Q, Han Y, Dy ABC, Hagerman RJ. The Gut Microbiota and Autism Spectrum Disorders. Front Cell Neurosci 2017; 11:120. [PMID: 28503135 PMCID: PMC5408485 DOI: 10.3389/fncel.2017.00120] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/10/2017] [Indexed: 12/19/2022] Open
Abstract
Gastrointestinal (GI) symptoms are a common comorbidity in patients with autism spectrum disorder (ASD), but the underlying mechanisms are unknown. Many studies have shown alterations in the composition of the fecal flora and metabolic products of the gut microbiome in patients with ASD. The gut microbiota influences brain development and behaviors through the neuroendocrine, neuroimmune and autonomic nervous systems. In addition, an abnormal gut microbiota is associated with several diseases, such as inflammatory bowel disease (IBD), ASD and mood disorders. Here, we review the bidirectional interactions between the central nervous system and the gastrointestinal tract (brain-gut axis) and the role of the gut microbiota in the central nervous system (CNS) and ASD. Microbiome-mediated therapies might be a safe and effective treatment for ASD.
Collapse
Affiliation(s)
- Qinrui Li
- Department of Pediatrics, Peking University First HospitalBeijing, China
| | - Ying Han
- Department of Pediatrics, Peking University First HospitalBeijing, China
| | - Angel Belle C Dy
- School of Medicine and Public Health, Ateneo de Manila UniversityQuezon City, Philippines
| | - Randi J Hagerman
- MIND Institute, University of California Davis Medical CenterSacramento, CA, USA.,Department of Pediatrics, University of California Davis Medical CenterSacramento, CA, USA
| |
Collapse
|
775
|
Safe S, Li X. Endocrine disruption: Relevance of experimental studies in female animals to human studies. CURRENT OPINION IN TOXICOLOGY 2017; 3:12-19. [DOI: 10.1016/j.cotox.2017.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
776
|
Herrema H, IJzerman RG, Nieuwdorp M. Emerging role of intestinal microbiota and microbial metabolites in metabolic control. Diabetologia 2017; 60:613-617. [PMID: 28013341 DOI: 10.1007/s00125-016-4192-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 12/07/2016] [Indexed: 01/04/2023]
Abstract
The role of the intestinal microbiota and microbial metabolites in the maintenance of host health and development of metabolic disease has gained significant attention over the past decade. Mechanistic insight revealing causality, however, is scarce. Work by Ussar and co-workers demonstrates that a complex interaction between microbiota, host genetics and environmental factors is involved in metabolic disease development in mice. In addition, Perry and co-workers show that the microbial metabolite acetate augments insulin resistance in rats. These studies underscore an important role of the microbiota in the development of obesity and symptoms of type 2 diabetes in rodents. If causality can be demonstrated in humans, development of novel diagnostic and therapeutic tools that target the gut microbiota will have high potential.
Collapse
Affiliation(s)
- Hilde Herrema
- Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1055 AZ, Amsterdam, the Netherlands.
| | - Richard G IJzerman
- Department of Internal Medicine, Free University Medical Center, Amsterdam, the Netherlands
| | - Max Nieuwdorp
- Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1055 AZ, Amsterdam, the Netherlands
- Department of Internal Medicine, Free University Medical Center, Amsterdam, the Netherlands
- Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
777
|
Tang WHW, Kitai T, Hazen SL. Gut Microbiota in Cardiovascular Health and Disease. Circ Res 2017; 120:1183-1196. [PMID: 28360349 PMCID: PMC5390330 DOI: 10.1161/circresaha.117.309715] [Citation(s) in RCA: 1062] [Impact Index Per Article: 132.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 02/07/2023]
Abstract
Significant interest in recent years has focused on gut microbiota-host interaction because accumulating evidence has revealed that intestinal microbiota play an important role in human health and disease, including cardiovascular diseases. Changes in the composition of gut microbiota associated with disease, referred to as dysbiosis, have been linked to pathologies such as atherosclerosis, hypertension, heart failure, chronic kidney disease, obesity, and type 2 diabetes mellitus. In addition to alterations in gut microbiota composition, the metabolic potential of gut microbiota has been identified as a contributing factor in the development of diseases. Recent studies revealed that gut microbiota can elicit a variety of effects on the host. Indeed, the gut microbiome functions like an endocrine organ, generating bioactive metabolites, that can impact host physiology. Microbiota interact with the host through many pathways, including the trimethylamine/trimethylamine N-oxide pathway, short-chain fatty acids pathway, and primary and secondary bile acids pathways. In addition to these metabolism-dependent pathways, metabolism-independent processes are suggested to also potentially contribute to cardiovascular disease pathogenesis. For example, heart failure-associated splanchnic circulation congestion, bowel wall edema, and impaired intestinal barrier function are thought to result in bacterial translocation, the presence of bacterial products in the systemic circulation and heightened inflammatory state. These are thought to also contribute to further progression of heart failure and atherosclerosis. The purpose of the current review is to highlight the complex interplay between microbiota, their metabolites, and the development and progression of cardiovascular diseases. We will also discuss the roles of gut microbiota in normal physiology and the potential of modulating intestinal microbial inhabitants as novel therapeutic targets.
Collapse
Affiliation(s)
- W H Wilson Tang
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (W.H.W.T., S.L.H.); Department of Cardiovascular Medicine, Heart and Vascular Institute (W.H.W.T., T.K.); and Center for Clinical Genomics, Cleveland Clinic, Cleveland OH (W.H.W.T.).
| | - Takeshi Kitai
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (W.H.W.T., S.L.H.); Department of Cardiovascular Medicine, Heart and Vascular Institute (W.H.W.T., T.K.); and Center for Clinical Genomics, Cleveland Clinic, Cleveland OH (W.H.W.T.)
| | - Stanley L Hazen
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (W.H.W.T., S.L.H.); Department of Cardiovascular Medicine, Heart and Vascular Institute (W.H.W.T., T.K.); and Center for Clinical Genomics, Cleveland Clinic, Cleveland OH (W.H.W.T.)
| |
Collapse
|
778
|
Remote Sensing between Liver and Intestine: Importance of Microbial Metabolites. ACTA ACUST UNITED AC 2017; 3:101-113. [PMID: 28983453 DOI: 10.1007/s40495-017-0087-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent technological advancements including metagenomics sequencing and metabolomics have allowed the discovery of critical functions of gut microbiota in obesity, malnutrition, neurological disorders, asthma, and xenobiotic metabolism. Classification of the human gut microbiome into distinct "enterotypes" has been proposed to serve as a new paradigm for understanding the interplay between microbial variation and human disease phenotypes, as many organs are affected by gut microbiota modifications during the pathogenesis of diseases. Gut microbiota remotely interacts with liver and other metabolic organs of the host through various microbial metabolites that are absorbed into the systemic circulation. PURPOSE OF REVIEW The present review summarizes recent literature regarding the importance of gut microbiota in modulating the physiological and pathological responses of various host organs, and describes the functions of the known microbial metabolites that are involved in this remote sensing process, with a primary focus on the gut microbiota-liver axis. RECENT FINDINGS Under physiological conditions, gut microbiota modulates the hepatic transcriptome, proteome, and metabolome, most notably down-regulating cytochrome P450 3a mediated xenobiotic metabolism. Gut microbiome also modulates the rhythmicity in liver gene expression, likely through microbial metabolites, such as butyrate and propionate that serve as epigenetic modifiers. Additionally, the production of host hormones such as primary bile acids and glucagon like peptide 1 is altered by gut microbiota to modify intermediary metabolism of the host. SUMMARY Dysregulation of gut microbiota is implicated in various liver diseases such as alcoholic liver disease, non-alcoholic steatohepatitis, liver cirrhosis, cholangitis, and liver cancer. Gut microbiota modifiers such as probiotics and prebiotics are increasingly recognized as novel therapeutic modalities for liver and other types of human diseases.
Collapse
|
779
|
Silverman MA, Konnikova L, Gerber JS. Impact of Antibiotics on Necrotizing Enterocolitis and Antibiotic-Associated Diarrhea. Gastroenterol Clin North Am 2017; 46:61-76. [PMID: 28164853 PMCID: PMC5314436 DOI: 10.1016/j.gtc.2016.09.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Antibiotic treatment alters the composition and metabolic function of the intestinal microbiota. These alterations may contribute to the pathogenesis of necrotizing enterocolitis (NEC) and antibiotic-associated diarrhea (AAD). Recent studies are beginning to unravel the contribution of specific groups of microbes and their metabolic pathways to these diseases. Probiotics or other microbiota-targeted therapies may provide effect strategies to prevent and treat NEC and AAD.
Collapse
Affiliation(s)
- Michael A. Silverman
- Division of Infectious Diseases, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Liza Konnikova
- Department of Pediatric and Newborn Medicine, Brigham and Women’s Hospital, Boston, MA 02115 and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Jeffrey S. Gerber
- Center for Pediatric Clinical Effectiveness, Division of Infectious Diseases, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104 and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
780
|
Abstract
The gut microbiota provides a range of ecologic, metabolic, and immunomodulatory functions relevant to health and well-being. The gut microbiota not only responds quickly to changes in diet, but this dynamic equilibrium may be managed to prevent and/or treat acute and chronic diseases. This article provides a working definition of the term "microbiome" and uses two examples of dietary interventions for the treatment of large bowel conditions to emphasize the links between diet and microbiome. There remains a need to develop a better functional understanding of the microbiota, if its management for clinical utility is to be fully realized.
Collapse
Affiliation(s)
- Nida Murtaza
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, 37 Kent St, Brisbane, Queensland 4102, Australia
| | - Páraic Ó Cuív
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, 37 Kent St, Brisbane, Queensland 4102, Australia
| | - Mark Morrison
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, 37 Kent St, Brisbane, Queensland 4102, Australia.
| |
Collapse
|
781
|
Ward TL, Knights D, Gale CA. Infant fungal communities: current knowledge and research opportunities. BMC Med 2017; 15:30. [PMID: 28190400 PMCID: PMC5304398 DOI: 10.1186/s12916-017-0802-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/24/2017] [Indexed: 12/16/2022] Open
Abstract
The microbes colonizing the infant gastrointestinal tract have been implicated in later-life disease states such as allergies and obesity. Recently, the medical research community has begun to realize that very early colonization events may be most impactful on future health, with the presence of key taxa required for proper immune and metabolic development. However, most studies to date have focused on bacterial colonization events and have left out fungi, a clinically important sub-population of the microbiota. A number of recent findings indicate the importance of host-associated fungi (the mycobiota) in adult and infant disease states, including acute infections, allergies, and metabolism, making characterization of early human mycobiota an important frontier of medical research. This review summarizes the current state of knowledge with a focus on factors influencing infant mycobiota development and associations between early fungal exposures and health outcomes. We also propose next steps for infant fungal mycobiome research, including longitudinal studies of mother-infant pairs while monitoring long-term health outcomes, further exploration of bacterium-fungus interactions, and improved methods and databases for mycobiome quantitation.
Collapse
Affiliation(s)
- Tonya L Ward
- Biotechnology Institute, University of Minnesota, Saint Paul, MN, USA
| | - Dan Knights
- Biotechnology Institute, University of Minnesota, Saint Paul, MN, USA.,Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Cheryl A Gale
- Department of Pediatrics, University of Minnesota, 2450 Riverside Ave, Minneapolis, MN, 55454, USA.
| |
Collapse
|
782
|
Bhatia M. Understanding toxicology: mechanisms and applications. Cell Biol Toxicol 2017; 33:1-4. [PMID: 27714471 DOI: 10.1007/s10565-016-9363-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 09/27/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Madhav Bhatia
- Department of Pathology, University of Otago, Christchurch, 2 Riccarton Avenue, PO Box 4345, Christchurch, 8140, New Zealand.
| |
Collapse
|
783
|
Affiliation(s)
- M. Ludwig
- Institut fuer Vegetative Physiologie; Charité Universitaetsmedizin Berlin; Berlin Germany
| | - A. Högner
- Institut fuer Vegetative Physiologie; Charité Universitaetsmedizin Berlin; Berlin Germany
| | - A. Patzak
- Institut fuer Vegetative Physiologie; Charité Universitaetsmedizin Berlin; Berlin Germany
| |
Collapse
|
784
|
Godkin A, Smith KA. Chronic infections with viruses or parasites: breaking bad to make good. Immunology 2017; 150:389-396. [PMID: 28009488 PMCID: PMC5343343 DOI: 10.1111/imm.12703] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 12/02/2016] [Accepted: 12/16/2016] [Indexed: 12/19/2022] Open
Abstract
Eukaryotic forms of life have been continually invaded by microbes and larger multicellular parasites, such as helminths. Over a billion years ago bacterial endosymbionts permanently colonized eukaryotic cells leading to recognized organelles with a distinct genetic lineage, such as mitochondria and chloroplasts. Colonization of our skin and mucosal surfaces with bacterial commensals is now known to be important for host health. However, the contribution of chronic virus and parasitic infections to immune homeostasis is being increasingly questioned. Persistent infection does not necessarily equate to exhibiting a chronic illness: healthy hosts (e.g. humans) have chronic viral and parasitic infections with no evidence of disease. Indeed, there are now examples of complex interactions between these microbes and hosts that seem to confer an advantage to the host at a particular time, suggesting that the relationship has progressed along an axis from parasitic to commensal to one of a mutualistic symbiosis. This concept is explored using examples from viruses and parasites, considering how the relationships may be not only detrimental but also beneficial to the human host.
Collapse
Affiliation(s)
- Andrew Godkin
- Division of Infection and Immunity, Cardiff University, Cardiff, Glamorgan, UK
| | - Katherine A Smith
- Division of Infection and Immunity, Cardiff University, Cardiff, Glamorgan, UK
| |
Collapse
|
785
|
Rajagopala SV, Vashee S, Oldfield LM, Suzuki Y, Venter JC, Telenti A, Nelson KE. The Human Microbiome and Cancer. Cancer Prev Res (Phila) 2017; 10:226-234. [PMID: 28096237 DOI: 10.1158/1940-6207.capr-16-0249] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/27/2016] [Accepted: 12/30/2016] [Indexed: 11/16/2022]
Abstract
Recent scientific advances have significantly contributed to our understanding of the complex connection between the microbiome and cancer. Our bodies are continuously exposed to microbial cells, both resident and transient, as well as their byproducts, including toxic metabolites. Circulation of toxic metabolites may contribute to cancer onset or progression at locations distant from where a particular microbe resides. Moreover, microbes may migrate to other locations in the human body and become associated with tumor development. Several case-control metagenomics studies suggest that dysbiosis in the commensal microbiota is also associated with inflammatory disorders and various cancer types throughout the body. Although the microbiome influences carcinogenesis through mechanisms independent of inflammation and immune system, the most recognizable link is between the microbiome and cancer via the immune system, as the resident microbiota plays an essential role in activating, training, and modulating the host immune response. Immunologic dysregulation is likely to provide mechanistic explanations as to how our microbiome influences cancer development and cancer therapies. In this review, we discuss recent developments in understanding the human gut microbiome's relationship with cancer and the feasibility of developing novel cancer diagnostics based on microbiome profiles. Cancer Prev Res; 10(4); 226-34. ©2017 AACR.
Collapse
Affiliation(s)
| | - Sanjay Vashee
- J. Craig Venter Institute (JCVI), Rockville, Maryland
| | | | - Yo Suzuki
- J. Craig Venter Institute (JCVI), Rockville, Maryland
| | - J Craig Venter
- J. Craig Venter Institute (JCVI), Rockville, Maryland.,Human Longevity, Inc., San Diego, California
| | - Amalio Telenti
- J. Craig Venter Institute (JCVI), Rockville, Maryland.,Human Longevity, Inc., San Diego, California
| | - Karen E Nelson
- J. Craig Venter Institute (JCVI), Rockville, Maryland. .,Human Longevity, Inc., San Diego, California
| |
Collapse
|
786
|
Studying Vertical Microbiome Transmission from Mothers to Infants by Strain-Level Metagenomic Profiling. mSystems 2017; 2:mSystems00164-16. [PMID: 28144631 PMCID: PMC5264247 DOI: 10.1128/msystems.00164-16] [Citation(s) in RCA: 310] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/21/2016] [Indexed: 12/28/2022] Open
Abstract
The gut microbiome becomes shaped in the first days of life and continues to increase its diversity during the first months. Links between the configuration of the infant gut microbiome and infant health are being shown, but a comprehensive strain-level assessment of microbes vertically transmitted from mother to infant is still missing. We collected fecal and breast milk samples from multiple mother-infant pairs during the first year of life and applied shotgun metagenomic sequencing followed by computational strain-level profiling. We observed that several specific strains, including those of Bifidobacterium bifidum, Coprococcus comes, and Ruminococcus bromii, were present in samples from the same mother-infant pair, while being clearly distinct from those carried by other pairs, which is indicative of vertical transmission. We further applied metatranscriptomics to study the in vivo gene expression of vertically transmitted microbes and found that transmitted strains of Bacteroides and Bifidobacterium species were transcriptionally active in the guts of both adult and infant. By combining longitudinal microbiome sampling and newly developed computational tools for strain-level microbiome analysis, we demonstrated that it is possible to track the vertical transmission of microbial strains from mother to infants and to characterize their transcriptional activity. Our work provides the foundation for larger-scale surveys to identify the routes of vertical microbial transmission and its influence on postinfancy microbiome development. IMPORTANCE Early infant exposure is important in the acquisition and ultimate development of a healthy infant microbiome. There is increasing support for the idea that the maternal microbial reservoir is a key route of microbial transmission, and yet much is inferred from the observation of shared species in mother and infant. The presence of common species, per se, does not necessarily equate to vertical transmission, as species exhibit considerable strain heterogeneity. It is therefore imperative to assess whether shared microbes belong to the same genetic variant (i.e., strain) to support the hypothesis of vertical transmission. Here we demonstrate the potential of shotgun metagenomics and strain-level profiling to identify vertical transmission events. Combining these data with metatranscriptomics, we show that it is possible not only to identify and track the fate of microbes in the early infant microbiome but also to investigate the actively transcribing members of the community. These approaches will ultimately provide important insights into the acquisition, development, and community dynamics of the infant microbiome.
Collapse
|
787
|
Anderson PO. Adverse Drug Reactions. Breastfeed Med 2016; 11:501-503. [PMID: 27732053 DOI: 10.1089/bfm.2016.0156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Philip O Anderson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California , San Diego, La Jolla, California
| |
Collapse
|
788
|
Abstract
PURPOSE OF REVIEW The rise in the prevalence of food allergy over the past decades has focused attention of factors that may impact disease development, most notably the gut microbiota. The gut microbial communities play a crucial role in promoting oral tolerance. Their alteration by such factors as Cesarean section delivery, diet and antibiotics may influence disease development. This review highlights recent progress in our understanding of the role of the gut microbiota in the development of food allergy. RECENT FINDINGS Food allergy is associated with alterations in the gut microbiota or dysbiosis early in life that may be predictive of disease persistence versus tolerance acquisition. Evidence for the benefits of adjunct therapy with probiotics for the prevention of food allergies and for potentiating oral immunotherapy remains circumstantial, with further studies needed to validate its use. Studies in murine models of food allergy suggest that microbial therapy with protolerogenic bacteria such as certain Clostridial species holds promise in future applications for prevention or therapy of food allergy. SUMMARY Progress in understanding the role of dysbiosis in food allergy and the factors that promote its development, such as antibiotic therapy, diet, modes of infant delivery, and environmental exposures, offer windows of opportunity for both preventive and therapeutic interventions to stem the rising tide of the food allergy epidemic.
Collapse
|
789
|
Interactions of Respiratory Viruses and the Nasal Microbiota during the First Year of Life in Healthy Infants. mSphere 2016; 1:mSphere00312-16. [PMID: 27904883 PMCID: PMC5120172 DOI: 10.1128/msphere.00312-16] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 10/29/2016] [Indexed: 11/20/2022] Open
Abstract
Respiratory viral infections are very frequent in infancy and of importance in acute and chronic disease development. Infections with human rhinovirus (HRV) are, e.g., associated with the later development of asthma. We found that only symptomatic HRV infections were associated with acute changes in the nasal microbiota, mainly characterized by a loss of microbial diversity. Infants with more frequent symptomatic HRV infections had a lower bacterial diversity at the end of the first year of life. Whether the interaction between viruses and the microbiota is one pathway contributing to asthma development will be assessed in the follow-ups of these children. Independent of that, measurements of microbial diversity might represent a potential marker for risk of later lung disease or monitoring of early life interventions. Traditional culture techniques have shown that increased bacterial colonization is associated with viral colonization; however, the influence of viral colonization on the whole microbiota composition is less clear. We thus aimed to understand the interaction of viral infections and the nasal microbiota in early life to appraise their roles in disease development. Thirty-two healthy, unselected infants were included in this prospective longitudinal cohort study within the first year of life. Biweekly nasal swabs (n = 559) were taken, and the microbiota was analyzed by 16S rRNA pyrosequencing, and 10 different viruses and 2 atypical bacteria were characterized by real-time PCR (combination of seven duplex samples). In contrast to asymptomatic human rhinovirus (HRV) colonization, symptomatic HRV infections were associated with lower alpha diversity (Shannon diversity index [SDI]), higher bacterial density (PCR concentration), and a difference in beta diversities (Jaccard and Bray-Curtis index) of the microbiota. In addition, infants with more frequent HRV infections had a lower SDI at the end of the study period. Overall, changes in the microbiota associated with symptomatic HRV infections were characterized by a loss of microbial diversity. The interaction between HRV infections and the nasal microbiota in early life might be of importance for later disease development and indicate a potential approach for future interventions. IMPORTANCE Respiratory viral infections are very frequent in infancy and of importance in acute and chronic disease development. Infections with human rhinovirus (HRV) are, e.g., associated with the later development of asthma. We found that only symptomatic HRV infections were associated with acute changes in the nasal microbiota, mainly characterized by a loss of microbial diversity. Infants with more frequent symptomatic HRV infections had a lower bacterial diversity at the end of the first year of life. Whether the interaction between viruses and the microbiota is one pathway contributing to asthma development will be assessed in the follow-ups of these children. Independent of that, measurements of microbial diversity might represent a potential marker for risk of later lung disease or monitoring of early life interventions.
Collapse
|
790
|
Moos WH, Pinkert CA, Irwin MH, Faller DV, Kodukula K, Glavas IP, Steliou K. Epigenetic Treatment of Persistent Viral Infections. Drug Dev Res 2016; 78:24-36. [PMID: 27761936 DOI: 10.1002/ddr.21366] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Preclinical Research Approximately 2,500 years ago, Hippocrates used the word herpes as a medical term to describe lesions that appeared to creep or crawl on the skin, advocating heat as a possible treatment. During the last 50 years, pharmaceutical research has made great strides, and therapeutic options have expanded to include small molecule antiviral agents, protease inhibitors, preventive vaccines for a handful of the papillomaviruses, and even cures for hepatitis C virus infections. However, effective treatments for persistent and recurrent viral infections, particularly the highly prevalent herpesviruses, continue to represent a significant unmet medical need, affecting the majority of the world's population. Exploring the population diversity of the human microbiome and the effects its compositional variances have on the immune system, health, and disease are the subjects of intense investigational research and study. Among the collection of viruses, bacteria, fungi, and single-cell eukaryotes that comprise the human microbiome, the virome has been grossly understudied relative to the influence it exerts on human pathophysiology, much as mitochondria have until recently failed to receive the attention they deserve, given their critical biomedical importance. Fortunately, cellular epigenetic machinery offers a wealth of druggable targets for therapeutic intervention in numerous disease indications, including those outlined above. With advances in synthetic biology, engineering our body's commensal microorganisms to seek out and destroy pathogenic species is clearly on the horizon. This is especially the case given recent breakthroughs in genetic manipulation with tools such as the CRISPR/Cas (clustered regularly interspaced short palindromic repeats/CRISPR-associated) gene-editing platforms. Tying these concepts together with our previous work on the microbiome and neurodegenerative and neuropsychiatric diseases, we suggest that, because mammalian cells respond to a viral infection by triggering a cascade of antiviral innate immune responses governed substantially by the cell's mitochondria, small molecule carnitinoids represent a new class of therapeutics with potential widespread utility against many infectious insults. Drug Dev Res 78 : 24-36, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Walter H Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, California
| | - Carl A Pinkert
- Department of Biological Sciences, College of Arts and Sciences, The University of Alabama, Tuscaloosa, Alabama
| | - Michael H Irwin
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Douglas V Faller
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts.,Boston University School of Medicine, Cancer Research Center, Boston, Massachusetts
| | | | - Ioannis P Glavas
- Department of Ophthalmology, New York University School of Medicine, New York
| | - Kosta Steliou
- Boston University School of Medicine, Cancer Research Center, Boston, Massachusetts.,PhenoMatriX, Boston, Massachusetts
| |
Collapse
|
791
|
Slattery J, MacFabe DF, Frye RE. The Significance of the Enteric Microbiome on the Development of Childhood Disease: A Review of Prebiotic and Probiotic Therapies in Disorders of Childhood. Clin Med Insights Pediatr 2016; 10:91-107. [PMID: 27774001 PMCID: PMC5063840 DOI: 10.4137/cmped.s38338] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 08/30/2016] [Accepted: 09/05/2016] [Indexed: 02/07/2023] Open
Abstract
Recent studies have highlighted the fact that the enteric microbiome, the trillions of microbes that inhabit the human digestive tract, has a significant effect on health and disease. Methods for manipulating the enteric microbiome, particularly through probiotics and microbial ecosystem transplantation, have undergone some study in clinical trials. We review some of the evidence for microbiome alteration in relation to childhood disease and discuss the clinical trials that have examined the manipulation of the microbiome in an effort to prevent or treat childhood disease with a primary focus on probiotics, prebiotics, and/or synbiotics (ie, probiotics + prebiotics). Studies show that alterations in the microbiome may be a consequence of events occurring during infancy and/or childhood such as prematurity, C-sections, and nosocomial infections. In addition, certain childhood diseases have been associated with microbiome alterations, namely necrotizing enterocolitis, infantile colic, asthma, atopic disease, gastrointestinal disease, diabetes, malnutrition, mood/anxiety disorders, and autism spectrum disorders. Treatment studies suggest that probiotics are potentially protective against the development of some of these diseases. Timing and duration of treatment, the optimal probiotic strain(s), and factors that may alter the composition and function of the microbiome are still in need of further research. Other treatments such as prebiotics, fecal microbial transplantation, and antibiotics have limited evidence. Future translational work, in vitro models, long-term and follow-up studies, and guidelines for the composition and viability of probiotic and microbial therapies need to be developed. Overall, there is promising evidence that manipulating the microbiome with probiotics early in life can help prevent or reduce the severity of some childhood diseases, but further research is needed to elucidate biological mechanisms and determine optimal treatments.
Collapse
Affiliation(s)
- John Slattery
- Arkansas Children’s Research Institute, Little Rock, AR, USA
- Division of Neurology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Derrick F. MacFabe
- The Kilee Patchell-Evans Autism Research Group, Departments of Psychology (Neuroscience) and Psychiatry, Division of Developmental Disabilities, University of Western Ontario, London, ON, Canada
| | - Richard E. Frye
- Arkansas Children’s Research Institute, Little Rock, AR, USA
- Division of Neurology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
792
|
Abstract
The ecosystem of the human gut consists of trillions of bacteria forming a bioreactor that is fueled by dietary macronutrients to produce bioactive compounds. These microbiota-derived metabolites signal to distant organs in the body, which enables the gut bacteria to connect to the immune and hormone system, to the brain (the gut-brain axis) and to host metabolism, as well as other functions of the host. This microbe-host communication is essential to maintain vital functions of the healthy host. Recently, however, the gut microbiota has been associated with a number of diseases, ranging from obesity and inflammatory diseases to behavioral and physiological abnormalities associated with neurodevelopmental disorders. In this Review, we will discuss microbiota-host cross-talk and intestinal microbiome signaling to extraintestinal organs. We will review mechanisms of how this communication might contribute to host physiology and discuss how misconfigured signaling might contribute to different diseases.
Collapse
|
793
|
Signals from the gut microbiota to distant organs in physiology and disease. Nat Med 2016; 22:1079-1089. [DOI: 10.1038/nm.4185] [Citation(s) in RCA: 695] [Impact Index Per Article: 77.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/23/2016] [Indexed: 02/06/2023]
|
794
|
Gasparrini AJ, Crofts TS, Gibson MK, Tarr PI, Warner BB, Dantas G. Antibiotic perturbation of the preterm infant gut microbiome and resistome. Gut Microbes 2016; 7:443-9. [PMID: 27472377 PMCID: PMC5154371 DOI: 10.1080/19490976.2016.1218584] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The gut microbiota plays important roles in nutrient absorption, immune system development, and pathogen colonization resistance. Perturbations early in life may be detrimental to host health in the short and the long-term. Antibiotics are among the many factors that influence the development of the microbiota. Because antibiotics are heavily administered during the first critical years of gut microbiota development, it is important to understand the effects of these interventions. Infants, particularly those born prematurely, represent an interesting population because they receive early and often extensive antibiotic therapy in the first months after birth. Gibson et al. recently demonstrated that antibiotic therapy in preterm infants can dramatically affect the gut microbiome. While meropenem, ticarcillin-clavulanate, and cefotaxime treatments were associated with decreased species richness, gentamicin and vancomycin had variable effects on species richness. Interestingly, the direction of species richness response could be predicted based on the abundance of 2 species and 2 genes in the microbiome prior to gentamicin or vancomycin treatment. Nonetheless, all antibiotic treatments enriched the presence of resistance genes and multidrug resistant organisms. Treatment with different antibiotics further resulted in unique population shifts of abundant organisms and selection for different sets of resistance genes. In this addendum, we provide an extended discussion of these recent findings, and outline important future directions for elucidating the interplay between antibiotics and preterm infant gut microbiota development.
Collapse
Affiliation(s)
- Andrew J. Gasparrini
- Center for Genome Sciences and Systems
Biology, Washington University School of Medicine, St Louis, MO,
USA
| | - Terence S. Crofts
- Center for Genome Sciences and Systems
Biology, Washington University School of Medicine, St Louis, MO,
USA,Department of Pathology and Immunology,
Washington University School of Medicine, St Louis, MO,
USA
| | - Molly K. Gibson
- Center for Genome Sciences and Systems
Biology, Washington University School of Medicine, St Louis, MO,
USA
| | - Phillip I. Tarr
- Department of Pediatrics, Washington
University School of Medicine, St Louis, MO, USA,Department of Molecular Microbiology,
Washington University School of Medicine, St Louis, MO,
USA
| | - Barbara B. Warner
- Department of Pediatrics, Washington
University School of Medicine, St Louis, MO, USA
| | - Gautam Dantas
- Center for Genome Sciences and Systems
Biology, Washington University School of Medicine, St Louis, MO,
USA,Department of Pathology and Immunology,
Washington University School of Medicine, St Louis, MO,
USA,Department of Molecular Microbiology,
Washington University School of Medicine, St Louis, MO,
USA,Department of Biomedical Engineering,
Washington University, St Louis, MO, USA
| |
Collapse
|