101
|
Park HJ, Lee SW, Kwon DJ, Heo SI, Park SH, Kim SY, Hong S. Oral administration of taheebo (Tabebuia avellanedae Lorentz ex Griseb.) water extract prevents DSS-induced colitis in mice by up-regulating type II T helper immune responses. Altern Ther Health Med 2017; 17:448. [PMID: 28877696 PMCID: PMC5585948 DOI: 10.1186/s12906-017-1952-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/30/2017] [Indexed: 02/07/2023]
Abstract
Background Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders that are mediated by pathogenic Th1 and Th17 cells. Previous studies have demonstrated that taheebo water extract (TWE) derived from Tabebuia avellanedae Lorentz ex Griseb., as folk remedy, has been used to treat various inflammatory diseases. Although TWE has been previously shown to display anti-inflammatory activities, the in vivo effects of TWE on mucosal immune responses remain unclear. Methods We examined the anti-inflammatory effects of TWE on innate immune cells such as dendritic cells (DCs) and macrophages and also on the differentiation of T helper cells. Lastly, adopting a method for dextran sulfate sodium (DSS)-induced colitis, we investigated whether the oral administration of TWE can modulate mucosal inflammatory responses. Results We found that TWE could activate DCs to produce immunosuppressive IL10 and polarize macrophages toward an anti-inflammatory phenotype in the mesenteric lymph node (MLN). Such alterations in DCs and macrophages resulted in a significant increase in anti-inflammatory Th2 and Foxp3+ Treg cells and a dramatic decrease in pro-inflammatory Th1 and Th17 cells in the MLN. Upon induction of colitis with DSS treatment, TWE significantly reduced the clinical symptoms, including body weight loss and colonic tissue inflammation, by up-regulating type II T helper immune responses. Conclusions Taken together, these data suggest that TWE is an excellent natural product with therapeutic effects to help improve inflammatory disorders such as colitis.
Collapse
|
102
|
Chabé M, Lokmer A, Ségurel L. Gut Protozoa: Friends or Foes of the Human Gut Microbiota? Trends Parasitol 2017; 33:925-934. [PMID: 28870496 DOI: 10.1016/j.pt.2017.08.005] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 07/17/2017] [Accepted: 08/11/2017] [Indexed: 12/18/2022]
Abstract
The importance of the gut microbiota for human health has sparked a strong interest in the study of the factors that shape its composition and diversity. Despite the growing evidence suggesting that helminths and protozoa significantly interact with gut bacteria, gut microbiome studies remain mostly focused on prokaryotes and on populations living in industrialized countries that typically have a low parasite burden. We argue that protozoa, like helminths, represent an important factor to take into account when studying the gut microbiome, and that their presence - especially considering their long coevolutionary history with humans - may be beneficial. From this perspective, we examine the relationship between the protozoa and their hosts, as well as their relevance for public health.
Collapse
Affiliation(s)
- Magali Chabé
- University of Lille, CNRS, Inserm, CHU de Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Ana Lokmer
- UMR 7206 Eco-anthropologie et ethnobiologie, CNRS - MNHN - Paris Diderot University - Sorbonne Paris Cité, Paris, France
| | - Laure Ségurel
- UMR 7206 Eco-anthropologie et ethnobiologie, CNRS - MNHN - Paris Diderot University - Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
103
|
Lippens C, Faivre B, Sorci G. Microevolutionary response of a gut nematode to intestinal inflammation. Int J Parasitol 2017; 47:617-623. [DOI: 10.1016/j.ijpara.2017.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/07/2017] [Accepted: 03/09/2017] [Indexed: 12/26/2022]
|
104
|
Case-Control Comparison of Enteric Viromes in Captive Rhesus Macaques with Acute or Idiopathic Chronic Diarrhea. J Virol 2017; 91:JVI.00952-17. [PMID: 28659484 DOI: 10.1128/jvi.00952-17] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 06/13/2017] [Indexed: 12/12/2022] Open
Abstract
Diarrhea is the major cause of non-research-associated morbidity and mortality affecting the supply of rhesus macaques and, potentially, their responses to experimental treatments. Idiopathic chronic diarrhea (ICD) in rhesus macaques also resembles ulcerative colitis, one form of human inflammatory bowel disease. To test for viral etiologies, we characterized and compared the fecal viromes from 32 healthy animals, 31 animals with acute diarrhea, and 29 animals with ICD. The overall fractions of eukaryotic viral reads were 0.063% for the healthy group, 0.131% for the acute-diarrhea group, and 0.297% for the chronic-diarrhea group. Eukaryotic viruses belonging to 6 viral families, as well as numerous circular Rep-encoding single-stranded DNA (CRESS DNA) viral genomes, were identified. The most commonly detected sequences were from picornaviruses, making up 59 to 88% of all viral reads, followed by 9 to 17% for CRESS DNA virus sequences. The remaining 5 virus families, Adenoviridae, Astroviridae, Anelloviridae, Picobirnaviridae, and Parvoviridae, collectively made up 1 to 3% of the viral reads, except for parvoviruses, which made up 23% of the viral reads in the healthy group. Detected members of the families Picornaviridae and Parvoviridae were highly diverse, consisting of multiple genera, species, and genotypes. Coinfections with members of up to six viral families were detected. Complete and partial viral genomes were assembled and used to measure the number of matching short sequence reads in feces from the 92 animals in the two clinical and the healthy control groups. Several enterovirus genotypes and CRESS DNA genomes were associated with ICD relative to healthy animals. Conversely, higher read numbers from different parvoviruses were associated with healthy animals. Our study reveals a high level of enteric coinfections with diverse viruses in a captive rhesus macaque colony and identifies several viruses positively or negatively associated with ICD.
Collapse
|
105
|
Wegener Parfrey L, Jirků M, Šíma R, Jalovecká M, Sak B, Grigore K, Jirků Pomajbíková K. A benign helminth alters the host immune system and the gut microbiota in a rat model system. PLoS One 2017; 12:e0182205. [PMID: 28771620 PMCID: PMC5542714 DOI: 10.1371/journal.pone.0182205] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 07/16/2017] [Indexed: 12/26/2022] Open
Abstract
Helminths and bacteria are major players in the mammalian gut ecosystem and each influences the host immune system and health. Declines in helminth prevalence and bacterial diversity appear to play a role in the dramatic rise of immune mediated inflammatory diseases (IMIDs) in western populations. Helminths are potent modulators of immune system and their reintroduction is a promising therapeutic avenue for IMIDs. However, the introduction of helminths represents a disturbance for the host and it is important to understand the impact of helminth reintroduction on the host, including the immune system and gut microbiome. We tested the impact of a benign tapeworm, Hymenolepis diminuta, in a rat model system. We find that H. diminuta infection results in increased interleukin 10 gene expression in the beginning of the prepatent period, consistent with induction of a type 2 immune response. We also find induction of humoral immunity during the patent period, shown here by increased IgA in feces. Further, we see an immuno-modulatory effect in the small intestine and spleen in patent period, as measured by reductions in tissue immune cells. We observed shifts in microbiota community composition during the patent period (beta-diversity) in response to H. diminuta infection. However, these compositional changes appear to be minor; they occur within families and genera common to both treatment groups. There was no change in alpha diversity. Hymenolepis diminuta is a promising model for helminth therapy because it establishes long-term, stable colonization in rats and modulates the immune system without causing bacterial dysbiosis. These results suggest that the goal of engineering a therapeutic helminth that can safely manipulate the mammalian immune system without disrupting the rest of the gut ecosystem is in reach.
Collapse
Affiliation(s)
- Laura Wegener Parfrey
- Departments of Botany and Zoology, University of British Columbia, Vancouver, Canada.,Integrated Microbial Biodiversity Program, Canadian Institute for Advanced Research, Toronto, Canada
| | - Milan Jirků
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Radek Šíma
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Marie Jalovecká
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Bohumil Sak
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Karina Grigore
- Departments of Botany and Zoology, University of British Columbia, Vancouver, Canada
| | - Kateřina Jirků Pomajbíková
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
| |
Collapse
|
106
|
Peachey LE, Jenkins TP, Cantacessi C. This Gut Ain’t Big Enough for Both of Us. Or Is It? Helminth–Microbiota Interactions in Veterinary Species. Trends Parasitol 2017; 33:619-632. [DOI: 10.1016/j.pt.2017.04.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/12/2017] [Accepted: 04/13/2017] [Indexed: 01/25/2023]
|
107
|
Varyani F, Fleming JO, Maizels RM. Helminths in the gastrointestinal tract as modulators of immunity and pathology. Am J Physiol Gastrointest Liver Physiol 2017; 312:G537-G549. [PMID: 28302598 PMCID: PMC5495915 DOI: 10.1152/ajpgi.00024.2017] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/06/2017] [Accepted: 03/12/2017] [Indexed: 01/31/2023]
Abstract
Helminth parasites are highly prevalent in many low- and middle-income countries, in which inflammatory bowel disease and other immunopathologies are less frequent than in the developed world. Many of the most common helminths establish themselves in the gastrointestinal tract and can exert counter-inflammatory influences on the host immune system. For these reasons, interest has arisen as to how parasites may ameliorate intestinal inflammation and whether these organisms, or products they release, could offer future therapies for immune disorders. In this review, we discuss interactions between helminth parasites and the mucosal immune system, as well as the progress being made toward identifying mechanisms and molecular mediators through which it may be possible to attenuate pathology in the intestinal tract.
Collapse
Affiliation(s)
- Fumi Varyani
- 1Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom; ,2Edinburgh Clinical Academic Track, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom; and
| | - John O. Fleming
- 3Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Rick M. Maizels
- 1Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom;
| |
Collapse
|
108
|
Tropini C, Earle KA, Huang KC, Sonnenburg JL. The Gut Microbiome: Connecting Spatial Organization to Function. Cell Host Microbe 2017; 21:433-442. [PMID: 28407481 DOI: 10.1016/j.chom.2017.03.010] [Citation(s) in RCA: 417] [Impact Index Per Article: 52.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The first rudimentary evidence that the human body harbors a microbiota hinted at the complexity of host-associated microbial ecosystems. Now, almost 400 years later, a renaissance in the study of microbiota spatial organization, driven by coincident revolutions in imaging and sequencing technologies, is revealing functional relationships between biogeography and health, particularly in the vertebrate gut. In this Review, we present our current understanding of principles governing the localization of intestinal bacteria, and spatial relationships between bacteria and their hosts. We further discuss important emerging directions that will enable progressing from the inherently descriptive nature of localization and -omics technologies to provide functional, quantitative, and mechanistic insight into this complex ecosystem.
Collapse
Affiliation(s)
- Carolina Tropini
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kristen A Earle
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kerwyn Casey Huang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| | | |
Collapse
|
109
|
Midha A, Schlosser J, Hartmann S. Reciprocal Interactions between Nematodes and Their Microbial Environments. Front Cell Infect Microbiol 2017; 7:144. [PMID: 28497029 PMCID: PMC5406411 DOI: 10.3389/fcimb.2017.00144] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/07/2017] [Indexed: 01/07/2023] Open
Abstract
Parasitic nematode infections are widespread in nature, affecting humans as well as wild, companion, and livestock animals. Most parasitic nematodes inhabit the intestines of their hosts living in close contact with the intestinal microbiota. Many species also have tissue migratory life stages in the absence of severe systemic inflammation of the host. Despite the close coexistence of helminths with numerous microbes, little is known concerning these interactions. While the environmental niche is considerably different, the free-living nematode Caenorhabditis elegans (C. elegans) is also found amongst a diverse microbiota, albeit on decaying organic matter. As a very well characterized model organism that has been intensively studied for several decades, C. elegans interactions with bacteria are much more deeply understood than those of their parasitic counterparts. The enormous breadth of understanding achieved by the C. elegans research community continues to inform many aspects of nematode parasitology. Here, we summarize what is known regarding parasitic nematode-bacterial interactions while comparing and contrasting this with information from work in C. elegans. This review highlights findings concerning responses to bacterial stimuli, antimicrobial peptides, and the reciprocal influences between nematodes and their environmental bacteria. Furthermore, the microbiota of nematodes as well as alterations in the intestinal microbiota of mammalian hosts by helminth infections are discussed.
Collapse
Affiliation(s)
- Ankur Midha
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität BerlinBerlin, Germany
| | - Josephine Schlosser
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität BerlinBerlin, Germany
| | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität BerlinBerlin, Germany
| |
Collapse
|
110
|
Tao L, Reese TA. Making Mouse Models That Reflect Human Immune Responses. Trends Immunol 2017; 38:181-193. [PMID: 28161189 DOI: 10.1016/j.it.2016.12.007] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/19/2016] [Accepted: 12/30/2016] [Indexed: 02/08/2023]
Abstract
Humans are infected with a variety of acute and chronic pathogens over the course of their lives, and pathogen-driven selection has shaped the immune system of humans. The same is likely true for mice. However, laboratory mice we use for most biomedical studies are bred in ultra-hygienic environments, and are kept free of specific pathogens. We review recent studies that indicate that pathogen infections are important for the basal level of activation and the function of the immune system. Consideration of these environmental exposures of both humans and mice can potentially improve mouse models of human disease.
Collapse
Affiliation(s)
- Lili Tao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tiffany A Reese
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
111
|
Hoffman DJ, Campos-Ponce M, Taddei CR, Doak CM. Microbiome, growth retardation and metabolism: are they related? Ann Hum Biol 2017; 44:201-207. [PMID: 27927018 DOI: 10.1080/03014460.2016.1267261] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CONTEXT Despite an improvement in food security and the delivery of nutritional supplements to children living in impoverished parts of the world, poor growth is still highly prevalent. Given that the microbiome is related to both nutrient absorption, as well as overweight/obesity, it may play a central role in limiting or modifying normal growth processes while contributing to chronic disease risks. OBJECTIVE The objective of this paper is to describe normal growth processes, the role of the microbiome in supporting or disrupting normal growth processes, and its potential impact on long-term health. METHODS A literature search of relevant human and laboratory research on growth, microbiome and the relationship between poor growth and chronic diseases was conducted. This review focuses on potential mechanisms that implicate the microbiome as a mediator of poor growth and later metabolic outcomes. In this relationship, attention was given to the potential for gastrointestinal infections to disrupt the microbiome. RESULTS Based on the studies reviewed, it is clear that exposure to infections disturbs both intestinal functioning as well as normal growth and changes in the microbiome may influence micronutrient availability and metabolic processes. CONCLUSIONS The microbiome may play a significant role in limiting human growth, but little is known about changes in the microbiome during periods of undernutrition. Thus, it is of great scientific and public health importance to improve the understanding of how the microbiome changes during nutrient deprivation. To best address the issue of the double burden and poor growth in low-income countries, research is warranted to advance the knowledge of the long-term role of the microbiome in the health of children exposed to undernutrition.
Collapse
Affiliation(s)
- Daniel J Hoffman
- a Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition, and Health , Rutgers, the State University of New Jersey , New Brunswick , NJ , USA
| | - Maiza Campos-Ponce
- b Department of Health Sciences , VU University , Amsterdam , The Netherlands
| | - Carla R Taddei
- c Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences , University of São Paulo , São Paulo, SP , Brazil.,d School of Arts, Science and Humanities , University of São Paulo , São Paulo, SP , Brazil
| | - Colleen M Doak
- b Department of Health Sciences , VU University , Amsterdam , The Netherlands
| |
Collapse
|
112
|
Fourie NH, Wang D, Abey SK, Creekmore AL, Hong S, Martin CG, Wiley JW, Henderson WA. Structural and functional alterations in the colonic microbiome of the rat in a model of stress induced irritable bowel syndrome. Gut Microbes 2017; 8:33-45. [PMID: 28059627 PMCID: PMC5341915 DOI: 10.1080/19490976.2016.1273999] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 11/29/2016] [Accepted: 12/13/2016] [Indexed: 02/03/2023] Open
Abstract
Stress is known to perturb the microbiome and exacerbate irritable bowel syndrome (IBS) associated symptoms. Characterizing structural and functional changes in the microbiome is necessary to understand how alterations affect the biomolecular environment of the gut in IBS. Repeated water avoidance (WA) stress was used to induce IBS-like symptoms in rats. The colon-mucosa associated microbiome was characterized in 13 stressed and control animals by 16S sequencing. In silico analysis of the functional domains of microbial communities was done by inferring metagenomic profiles from 16S data. Microbial communities and functional profiles were compared between conditions. WA animals exhibited higher α-diversity and moderate divergence in community structure (β-diversity) compared with controls. Specific clades and taxa were consistently and significantly modified in the WA animals. The WA microbiome was particularly enriched in Proteobacteria and depleted in several beneficial taxa. A decreased capacity in metabolic domains, including energy- and lipid-metabolism, and an increased capacity for fatty acid and sulfur metabolism was inferred for the WA microbiome. The stressed condition favored the proliferation of a greater diversity of microbes that appear to be functionally similar, resulting in a functionally poorer microbiome with implications for epithelial health. Taxa, with known beneficial effects, were found to be depleted, which supports their relevance as therapeutic agents to restore microbial health. Microbial sulfur metabolism may form a key component of visceral nerve sensitization pathways and is therefore of interest as a target metabolic domain in microbial ecological restoration.
Collapse
Affiliation(s)
- Nicolaas H Fourie
- a National Institutes of Health, Division of Intramural Research, NINR, DHHS , Bethesda , MD , USA
| | - Dan Wang
- a National Institutes of Health, Division of Intramural Research, NINR, DHHS , Bethesda , MD , USA
| | - Sarah K Abey
- a National Institutes of Health, Division of Intramural Research, NINR, DHHS , Bethesda , MD , USA
| | - Amy L Creekmore
- b University of Michigan Medical School , Department of Internal Medicine - Gastroenterology , Ann Arbor , MI , USA
| | - Shuangsong Hong
- b University of Michigan Medical School , Department of Internal Medicine - Gastroenterology , Ann Arbor , MI , USA
| | - Christiana G Martin
- a National Institutes of Health, Division of Intramural Research, NINR, DHHS , Bethesda , MD , USA
| | - John W Wiley
- b University of Michigan Medical School , Department of Internal Medicine - Gastroenterology , Ann Arbor , MI , USA
| | - Wendy A Henderson
- a National Institutes of Health, Division of Intramural Research, NINR, DHHS , Bethesda , MD , USA
| |
Collapse
|
113
|
|
114
|
Carlsson AM, Mastromonaco G, Vandervalk E, Kutz S. Parasites, stress and reindeer: infection with abomasal nematodes is not associated with elevated glucocorticoid levels in hair or faeces. CONSERVATION PHYSIOLOGY 2016; 4:cow058. [PMID: 27957334 PMCID: PMC5147723 DOI: 10.1093/conphys/cow058] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 10/28/2016] [Accepted: 11/13/2016] [Indexed: 05/30/2023]
Abstract
Stress hormones (glucocorticoids), incorporated into hair/fur and faeces, have been proposed as biomarkers of overall health in wildlife. Although such biomarkers may be helpful for wildlife conservation and management, their use has rarely been validated. There is a paucity of studies examining the variation of stress hormones in mammals and how they relate to other health measures, such as parasitism. Parasites are ubiquitous in wildlife and can influence the fitness of individual animals and populations. Through a longitudinal experiment using captive reindeer (Rangifer tarandus tarandus), we tested whether animals infected with Ostertagia gruehneri, a gastrointestinal nematode with negative impacts on fitness of the host, had higher stress levels compared with those that had been treated to remove infection. Faecal samples were collected weekly for 12 weeks (June-September) and hair was collected at the start and end of the study; glucocorticoids were quantified using enzyme immunoassays. Contrary to what was expected, infected reindeer had similar levels of cortisol in hair and slightly lower glucocorticoid metabolites in faeces compared with uninfected reindeer. Faecal corticosterone levels were higher than faecal cortisol levels, and only corticosterone increased significantly after a handling event. These results suggest that reindeer may use a tolerance strategy to cope with gastrointestinal nematodes and raise the question as to whether moderate infection intensities with nematodes are beneficial to the host. By removing nematodes we may have altered the gut microbiota, leading to the observed elevated faecal glucocorticoid metabolite levels in the treated reindeer. These findings demonstrate the importance of considering both cortisol and corticosterone in physiological studies, as there is mounting evidence that they may have different functionalities.
Collapse
Affiliation(s)
- A. M. Carlsson
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, Alberta, CanadaT2N 4Z6
| | - G. Mastromonaco
- Reproductive Physiology Unit, Toronto Zoo, 361A Old Finch Avenue, Scarborough, Ontario,CanadaM1B 5K7
| | - E. Vandervalk
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, Alberta, CanadaT2N 4Z6
| | - S. Kutz
- Canadian Cooperative Wildlife Health Centre Alberta, 3280 Hospital Drive NW, Calgary, AB T2N4Z6, Canada
| |
Collapse
|
115
|
Duarte AM, Jenkins TP, Latrofa MS, Giannelli A, Papadopoulos E, de Carvalho LM, Nolan MJ, Otranto D, Cantacessi C. Helminth infections and gut microbiota - a feline perspective. Parasit Vectors 2016; 9:625. [PMID: 27912797 PMCID: PMC5135779 DOI: 10.1186/s13071-016-1908-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/22/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Investigations of the relationships between the gut microbiota and gastrointestinal parasitic nematodes are attracting growing interest by the scientific community, driven by the need to better understand the contribution of parasite-associated changes in the composition of the gut flora to both host malnutrition and immune modulation. These studies have however been carried out mainly in humans and experimental animals, while knowledge of the make-up of the gut commensal flora in presence or absence of infection by parasitic nematodes in domestic animals is limited. In this study, we investigate the qualitative and quantitative impact that infections by a widespread parasite of cats (i.e. Toxocara cati) exert on the gut microbiota of feline hosts. METHODS The faecal microbiota of cats with patent infection by T. cati (= Tc+), as well as that of negative controls (= Tc-) was examined via high-throughput sequencing of the V3-V4 hypervariable region of the bacterial 16S rRNA gene, followed by bioinformatics and biostatistical analyses of sequence data. RESULTS A total of 2,325,366 useable high-quality sequences were generated from the faecal samples analysed in this study and subjected to further bioinformatics analyses, which led to the identification of 128 OTUs and nine bacterial phyla, respectively. The phylum Firmicutes was predominant in all samples analysed (mean of 53.0%), followed by the phyla Proteobacteria (13.8%), Actinobacteria (13.7%) and Bacteroidetes (10.1%). Among others, bacteria of the order Lactobacillales, the family Enterococcaceae and genera Enterococcus and Dorea showed a trend towards increased abundance in Tc+ compared with Tc- samples, while no significant differences in OTU richness and diversity were recorded between Tc+ and Tc- samples (P = 0.485 and P = 0.581, respectively). However, Canonical Correlation and Redundancy Analyses were able to separate samples by infection status (P = 0.030 and P = 0.015, respectively), which suggests a correlation between the latter and the composition of the feline faecal microbiota. CONCLUSIONS In spite of the relatively small number of samples analysed, subtle differences in the composition of the gut microbiota of Tc+ vs Tc- cats could be identified, some of which in accordance with current data from humans and laboratory animal hosts. Nevertheless, the findings from this study contribute valuable knowledge to the yet little explored area of parasite-microbiota interactions in domestic animals.
Collapse
Affiliation(s)
- Ana M. Duarte
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
- CIISA, Faculty of Veterinary Medicine, Universidade de Lisboa, Lisboa, Portugal
| | - Timothy P. Jenkins
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Maria S. Latrofa
- Department of Veterinary Medicine, University of Bari, Valenzano, Italy
| | - Alessio Giannelli
- Department of Veterinary Medicine, University of Bari, Valenzano, Italy
| | - Elias Papadopoulos
- School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Matthew J. Nolan
- Department of Pathology and Pathogen Biology, Royal Veterinary College, University of London, Hatfield, UK
| | - Domenico Otranto
- Department of Veterinary Medicine, University of Bari, Valenzano, Italy
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
116
|
Hand TW, Vujkovic-Cvijin I, Ridaura VK, Belkaid Y. Linking the Microbiota, Chronic Disease, and the Immune System. Trends Endocrinol Metab 2016; 27:831-843. [PMID: 27623245 PMCID: PMC5116263 DOI: 10.1016/j.tem.2016.08.003] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/08/2016] [Accepted: 08/09/2016] [Indexed: 12/11/2022]
Abstract
Chronic inflammatory diseases (CIDs) are the most important causes of mortality in the world today and are on the rise. We now know that immune-driven inflammation is critical in the etiology of these diseases, though the environmental triggers and cellular mechanisms that lead to their development are still mysterious. Many CIDs are associated with significant shifts in the microbiota toward inflammatory configurations, which can affect the host both by inducing local and systemic inflammation and by alterations in microbiota-derived metabolites. This review discusses recent findings suggesting that shifts in the microbiota may contribute to chronic disease via effects on the immune system.
Collapse
Affiliation(s)
- Timothy W. Hand
- R.K. Mellon Institute for Pediatric Research, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA, 15224
- Correspondence addressed to: Timothy Hand () or Yasmine Belkaid ()
| | - Ivan Vujkovic-Cvijin
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID/NIH, Bethesda, Maryland 20892, USA
| | - Vanessa K. Ridaura
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID/NIH, Bethesda, Maryland 20892, USA
| | - Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID/NIH, Bethesda, Maryland 20892, USA
- National Institute of Allergy and Infectious diseases (NIAID) Microbiome Program, National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| |
Collapse
|
117
|
Changes in duodenal tissue-associated microbiota following hookworm infection and consecutive gluten challenges in humans with coeliac disease. Sci Rep 2016; 6:36797. [PMID: 27827438 PMCID: PMC5101533 DOI: 10.1038/srep36797] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 10/21/2016] [Indexed: 12/26/2022] Open
Abstract
A reduced diversity of the gastrointestinal commensal microbiota is associated with the development of several inflammatory diseases. Recent reports in humans and animal models have demonstrated the beneficial therapeutic effects of infections by parasitic worms (helminths) in some inflammatory disorders, such as inflammatory bowel disease (IBD) and coeliac disease (CeD). Interestingly, these studies have described how helminths may alter the intestinal microbiota, potentially representing a mechanism by which they regulate inflammation. However, for practical reasons, these reports have primarily analysed the faecal microbiota. In the present investigation, we have assessed, for the first time, the changes in the microbiota at the site of infection by a parasitic helminth (hookworm) and gluten-dependent inflammation in humans with CeD using biopsy tissue from the duodenum. Hookworm infection and gluten exposure were associated with an increased abundance of species
within the Bacteroides phylum, as well as increases in the richness and diversity of the tissue-resident microbiota within the intestine, results that are consistent with previous reports using other helminth species in humans and animal models. Hence, this may represent a mechanism by which parasitic helminths may restore intestinal immune homeostasis and exert a therapeutic benefit in CeD, and potentially other inflammatory disorders.
Collapse
|
118
|
Dietary Gluten-Induced Gut Dysbiosis Is Accompanied by Selective Upregulation of microRNAs with Intestinal Tight Junction and Bacteria-Binding Motifs in Rhesus Macaque Model of Celiac Disease. Nutrients 2016; 8:nu8110684. [PMID: 27801835 PMCID: PMC5133072 DOI: 10.3390/nu8110684] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/12/2016] [Accepted: 10/18/2016] [Indexed: 12/15/2022] Open
Abstract
The composition of the gut microbiome reflects the overall health status of the host. In this study, stool samples representing the gut microbiomes from 6 gluten-sensitive (GS) captive juvenile rhesus macaques were compared with those from 6 healthy, age- and diet-matched peers. A total of 48 samples representing both groups were studied using V4 16S rRNA gene DNA analysis. Samples from GS macaques were further characterized based on type of diet administered: conventional monkey chow, i.e., wheat gluten-containing diet (GD), gluten-free diet (GFD), barley gluten-derived diet (BOMI) and reduced gluten barley-derived diet (RGB). It was hypothesized that the GD diet would lower the gut microbial diversity in GS macaques. This is the first report illustrating the reduction of gut microbial alpha-diversity (p < 0.05) following the consumption of dietary gluten in GS macaques. Selected bacterial families (e.g., Streptococcaceae and Lactobacillaceae) were enriched in GS macaques while Coriobacteriaceae was enriched in healthy animals. Within several weeks after the replacement of the GD by the GFD diet, the composition (beta-diversity) of gut microbiome in GS macaques started to change (p = 0.011) towards that of a normal macaque. Significance for alpha-diversity however, was not reached by the day 70 when the feeding experiment ended. Several inflammation-associated microRNAs (miR-203, -204, -23a, -23b and -29b) were upregulated (p < 0.05) in jejunum of 4 biopsied GS macaques fed GD with predicted binding sites on 16S ribosomal RNA of Lactobacillus reuteri (accession number: NR_025911), Prevotella stercorea (NR_041364) and Streptococcus luteciae (AJ297218) that were overrepresented in feces. Additionally, claudin-1, a validated tight junction protein target of miR-29b was significantly downregulated in jejunal epithelium of GS macaques. Taken together, we predict that with the introduction of effective treatments in future studies the diversity of gut microbiomes in GS macaques will approach those of healthy individuals. Further studies are needed to elucidate the regulatory pathways of inflammatory miRNAs in intestinal mucosa of GS macaques and to correlate their expression with gut dysbiosis.
Collapse
|
119
|
Wood JR, Perry GLW, Wilmshurst JM. Using palaeoecology to determine baseline ecological requirements and interaction networks for de‐extinction candidate species. Funct Ecol 2016. [DOI: 10.1111/1365-2435.12773] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Jamie R. Wood
- Long‐term Ecology Lab Landcare Research PO Box 69040 Lincoln7640 New Zealand
| | - George L. W. Perry
- School of Environment The University of Auckland Private Bag 92019 Auckland1142 New Zealand
| | - Janet M. Wilmshurst
- Long‐term Ecology Lab Landcare Research PO Box 69040 Lincoln7640 New Zealand
- School of Environment The University of Auckland Private Bag 92019 Auckland1142 New Zealand
| |
Collapse
|
120
|
Filyk HA, Osborne LC. The Multibiome: The Intestinal Ecosystem's Influence on Immune Homeostasis, Health, and Disease. EBioMedicine 2016; 13:46-54. [PMID: 27863931 PMCID: PMC5264270 DOI: 10.1016/j.ebiom.2016.10.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/05/2016] [Accepted: 10/05/2016] [Indexed: 12/13/2022] Open
Abstract
Mammalian evolution has occurred in the presence of mutualistic, commensal, and pathogenic micro- and macro-organisms for millennia. The presence of these organisms during mammalian evolution has allowed for intimate crosstalk between these colonizing species and the host immune system. In this review, we introduce the concept of the ‘multibiome’ to holistically refer to the biodiverse collection of bacteria, viruses, fungi and multicellular helminthic worms colonizing the mammalian intestine. Furthermore, we discuss new insights into multibiome-host interactions in the context of host-protective immunity and immune-mediated diseases, including inflammatory bowel disease and multiple sclerosis. Finally, we provide reasons to account for the multibiome in experimental design, analysis and in therapeutic applications. The intestinal multibiome is composed of bacteria, viruses, fungi, and eukaryotes. Mammals evolved alongside a complex and biodiverse multibiome. Cross-talk between the multibiome and the host regulates immunity and inflammation.
Collapse
Affiliation(s)
- Heather A Filyk
- Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Lisa C Osborne
- Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
121
|
Abstract
By reputation, the parasite is a pariah, an unwelcome guest. Infection with helminth parasites evokes stereotypic immune responses in humans and mice that are dominated by T helper (Th)-2 responses; thus, a hypothesis arises that infection with helminths would limit immunopathology in concomitant inflammatory disease. Although infection with some species of helminths can cause devastating disease and affect the course of microbial infections, analyses of rodent models of inflammatory disease reveal that infection with helminth parasites, or treatment with helminth extracts, can limit the severity of autoinflammatory disease, including colitis. Intriguing, but fewer, studies show that adoptive transfer of myeloid immune cells treated with helminth products/extracts in vitro can suppress inflammation. Herein, 3 facets of helminth therapy are reviewed and critiqued: treatment with viable ova or larvae, treatment with crude extracts of the worm or purified molecules, and cellular immunotherapy. The beneficial effect of helminth therapy often converges on the mobilization of IL-10 and regulatory/alternatively activated macrophages, while there are reports on transforming growth factor (TGF)-β, regulatory T cells and dendritic cells, and recent data suggest that helminth-evoked changes in the microbiota should be considered when defining anticolitic mechanisms. We speculate that if the data from animal models translate to humans, noting the heterogeneity therein, then the choice between use of viable helminth ova, helminth extracts/molecules or antigen-pulsed immune cells could be matched to disease management in defined cohorts of patients with inflammatory bowel disease.
Collapse
|
122
|
Iqbal S, Quigley EMM. Progress in Our Understanding of the Gut Microbiome: Implications for the Clinician. Curr Gastroenterol Rep 2016; 18:49. [PMID: 27448618 DOI: 10.1007/s11894-016-0524-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The investigation of the role of the microbial communities of our gastrointestinal tract (microbiota) has accelerated dramatically in recent years thanks to rapid developments in the technologies that allow us to fully enumerate and evaluate the full complement of bacterial species and strains that normally inhabit the gut. Laboratory studies in a range of inventive animal models continue to provide insights into the role of the microbiota in health and to generate plausible hypotheses relating to its potential involvement in the pathogenesis of human disease. Studies of the composition of human gut microbiota continue to accumulate but their interpretation needs to be tempered by an appreciation of the limitations of single-point-in-time studies of fecal samples from small study populations. Nevertheless, clinically important examples of a central role for microbiota-host interactions in disease pathogenesis have emerged and many more have been postulated but await confirmation in appropriately powered and conducted studies.
Collapse
Affiliation(s)
- Sara Iqbal
- Department of Medicine, Houston Methodist Hospital and Weill Cornell Medical College, 6550 Fannin St, SM 1001, Houston, TX, 77030, USA
| | - Eamonn M M Quigley
- Department of Medicine, Houston Methodist Hospital and Weill Cornell Medical College, 6550 Fannin St, SM 1001, Houston, TX, 77030, USA.
- David M. and Lynda K. Underwood Center for Digestive Disorders, Division of Gastroenterology and Hepatology, Houston Methodist Hospital and Weill Cornell Medical College, 6550 Fannin St, SM 1201, Houston, TX, 77030, USA.
| |
Collapse
|
123
|
Zaiss MM, Harris NL. Interactions between the intestinal microbiome and helminth parasites. Parasite Immunol 2016; 38:5-11. [PMID: 26345715 PMCID: PMC5019230 DOI: 10.1111/pim.12274] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 09/01/2015] [Indexed: 12/14/2022]
Abstract
Throughout evolution, both helminths and bacteria have inhabited our intestines. As intestinal helminths and bacteria inhabit the same environmental niche, it is likely that these organisms interact with, and impact on, each other. In addition, intestinal helminths are well known to alter intestinal physiology, permeability, mucous secretion and the production of antimicrobial peptides – all of which may impact on bacterial survival and spatial organization. Yet despite rapid advances in our understanding of host–intestinal bacteria interactions, the impact of helminths on this relationship has remained largely unexplored. Moreover, although intestinal helminths are generally accepted to possess potent immuno‐modulatory activity, it is unknown whether this capacity requires interactions with intestinal bacteria. We propose that this ‘ménage à trois’ situation is likely to have exerted a strong selective pressure on the development of our metabolic and immune systems. Whilst such pressures remain in developing countries, the eradication of helminths in industrialized countries has shifted this evolutionary balance, possibly underlying the increased development of chronic inflammatory diseases. Thus, helminth–bacteria interactions may represent a key determinant of healthy homoeostasis.
Collapse
Affiliation(s)
- M M Zaiss
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - N L Harris
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
124
|
Aberrant immune response with consequent vascular and connective tissue remodeling - causal to scleroderma and associated syndromes such as Raynaud phenomenon and other fibrosing syndromes? Curr Opin Rheumatol 2016; 28:571-6. [PMID: 27548652 DOI: 10.1097/bor.0000000000000333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Scleroderma and other autoimmune-induced connective tissue diseases are characterized by dysfunctions in the immune system, connective tissue and the vasculature. We are focusing on systemic sclerosis (SSc)-associated pulmonary hypertension, which remains a leading cause of death with only a 50-60% of 2-year survival rate. RECENT FINDINGS Much research and translational efforts have been directed at understanding the immune response that causes SSc and the networked interactions with the connective tissue and the vasculature. One of the unexpected findings was that in some cases the pathogenic immune response in SSc resembles the immune response to helminth parasites. During coevolution, means of communication were developed which protect the host from over-colonization with parasites and which protect the parasite from excessive host responses. One explanation for the geographically clustered occurrence of SSc is that environmental exposures combined with genetic predisposition turn on triggers of molecular and cellular modules that were once initiated by parasites. SUMMARY Future research is needed to further understand the parasite-derived signals that dampen the host response. Therapeutic helminth infection or treatment with parasite-derived response modifiers could be promising new management tools for autoimmune connective tissue diseases.
Collapse
|
125
|
Loke P, Lim YAL. Can Helminth Infection Reverse Microbial Dysbiosis? Trends Parasitol 2016; 31:534-535. [PMID: 26604162 DOI: 10.1016/j.pt.2015.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 10/02/2015] [Indexed: 11/19/2022]
Abstract
There is growing interest in treating inflammatory conditions with helminth infection. Recently, Loukas and colleagues have reported promising results from using experimental hookworm infection to reduce gluten sensitivity in celiac disease patients. Analysis of microbiota samples from the trial is contributing to our understanding of the complexity underlying helminth–microbiota–host relationships.
Collapse
Affiliation(s)
- P'ng Loke
- Department of Microbiology, New York University School of Medicine, New York, NY, USA.
| | - Yvonne A L Lim
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
126
|
Bono-Lunn D, Villeneuve C, Abdulhay NJ, Harker M, Parker W. Policy and regulations in light of the human body as a ‘superorganism’ containing multiple, intertwined symbiotic relationships. ACTA ACUST UNITED AC 2016. [DOI: 10.1080/10601333.2016.1210159] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
127
|
Cattadori IM, Sebastian A, Hao H, Katani R, Albert I, Eilertson KE, Kapur V, Pathak A, Mitchell S. Impact of Helminth Infections and Nutritional Constraints on the Small Intestine Microbiota. PLoS One 2016; 11:e0159770. [PMID: 27438701 PMCID: PMC4954658 DOI: 10.1371/journal.pone.0159770] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 07/06/2016] [Indexed: 01/08/2023] Open
Abstract
Helminth infections and nutrition can independently alter the composition and abundance of the gastrointestinal microbiota, however, their combined effect is poorly understood. Here, we used the T. retortaeformis-rabbit system to examine how the helminth infection and host restriction from coprophagy/ready-to-absorb nutrients affected the duodenal microbiota, and how these changes related to the acquired immune response at the site of infection. A factorial experiment was performed where the bacterial community, its functionality and the immune response were examined in four treatments (Infect, Infect+Collar, Control+Collar and Control). Helminths reduced the diversity and abundance of the microbiota while the combination of parasites and coprophagic restriction led to a more diversified and abundant microbiota than infected cases, without significantly affecting the intensity of infection. Animals restricted from coprophagy and free from parasites exhibited the richest and most abundant bacterial community. By forcing the individuals to absorb nutrients from less digested food, the coprophagic restriction appears to have facilitated the diversity and proliferation of bacteria in the duodenum. Changes in the microbiota were more clearly associated with changes in the immune response for the infected than the nutrient restricted animals. The functional and metabolic characteristics of the duodenal microbiota were not significantly different between treatments. Overall, infection and diet affect the gut microbiota but their interactions and outcome can be complex. These findings can have important implications for the development of control measures to helminth infections where poor nutrition/malnutrition can also be a concern.
Collapse
Affiliation(s)
- Isabella M. Cattadori
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, 16082 PA, United States of America
- Department of Biology, The Pennsylvania State University, University Park, 16082 PA, United States of America
- * E-mail:
| | - Aswathy Sebastian
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, 16082 PA, United States of America
| | - Han Hao
- Department of Statistics, The Pennsylvania State University, University Park, 16082 PA, United States of America
| | - Robab Katani
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, 16082 PA, United States of America
| | - Istvan Albert
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, 16082 PA, United States of America
| | - Kirsten E. Eilertson
- Department of Statistics, The Pennsylvania State University, University Park, 16082 PA, United States of America
| | - Vivek Kapur
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, 16082 PA, United States of America
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, 16082 PA, United States of America
| | - Ashutosh Pathak
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, 16082 PA, United States of America
- Department of Biology, The Pennsylvania State University, University Park, 16082 PA, United States of America
| | - Susan Mitchell
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, 16082 PA, United States of America
- Department of Biology, The Pennsylvania State University, University Park, 16082 PA, United States of America
| |
Collapse
|
128
|
Giacomin P, Agha Z, Loukas A. Helminths and Intestinal Flora Team Up to Improve Gut Health. Trends Parasitol 2016; 32:664-666. [PMID: 27234811 DOI: 10.1016/j.pt.2016.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 05/12/2016] [Indexed: 01/02/2023]
Abstract
Inflammatory bowel diseases (IBD) are associated with impaired intestinal barrier function, chronic inflammation, and microbial dysbiosis. In a recent publication in Science, Ramanan et al. used murine and human studies to demonstrate that infections with gastrointestinal helminths can protect against IBD by provoking immune responses that alter the balance of commensal and pathogenic bacteria in the intestine.
Collapse
Affiliation(s)
- Paul Giacomin
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.
| | - Zainab Agha
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
129
|
Audebert C, Even G, Cian A, Loywick A, Merlin S, Viscogliosi E, Chabé M. Colonization with the enteric protozoa Blastocystis is associated with increased diversity of human gut bacterial microbiota. Sci Rep 2016; 6:25255. [PMID: 27147260 PMCID: PMC4857090 DOI: 10.1038/srep25255] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/13/2016] [Indexed: 12/16/2022] Open
Abstract
Alterations in the composition of commensal bacterial populations, a phenomenon known as dysbiosis, are linked to multiple gastrointestinal disorders, such as inflammatory bowel disease and irritable bowel syndrome, or to infections by diverse enteric pathogens. Blastocystis is one of the most common single-celled eukaryotes detected in human faecal samples. However, the clinical significance of this widespread colonization remains unclear, and its pathogenic potential is controversial. To address the issue of Blastocystis pathogenicity, we investigated the impact of colonization by this protist on the composition of the human gut microbiota. For that purpose, we conducted a cross-sectional study including 48 Blastocystis-colonized patients and 48 Blastocystis-free subjects and performed an Ion Torrent 16S rDNA gene sequencing to decipher the Blastocystis-associated gut microbiota. Here, we report a higher bacterial diversity in faecal microbiota of Blastocystis colonized patients, a higher abundance of Clostridia as well as a lower abundance of Enterobacteriaceae. Our results contribute to suggesting that Blastocystis colonization is usually associated with a healthy gut microbiota, rather than with gut dysbiosis generally observed in metabolic or infectious inflammatory diseases of the lower gastrointestinal tract.
Collapse
Affiliation(s)
- Christophe Audebert
- GENES DIFFUSION, Douai, France.,PEGASE-Biosciences, Institut Pasteur de Lille, Lille, France
| | - Gaël Even
- GENES DIFFUSION, Douai, France.,PEGASE-Biosciences, Institut Pasteur de Lille, Lille, France
| | - Amandine Cian
- Univ. Lille, CNRS, Inserm, CHU de Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | | | - Alexandre Loywick
- GENES DIFFUSION, Douai, France.,PEGASE-Biosciences, Institut Pasteur de Lille, Lille, France
| | - Sophie Merlin
- GENES DIFFUSION, Douai, France.,PEGASE-Biosciences, Institut Pasteur de Lille, Lille, France
| | - Eric Viscogliosi
- Univ. Lille, CNRS, Inserm, CHU de Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Magali Chabé
- Univ. Lille, CNRS, Inserm, CHU de Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
130
|
Oriá RB, Murray-Kolb LE, Scharf RJ, Pendergast LL, Lang DR, Kolling GL, Guerrant RL. Early-life enteric infections: relation between chronic systemic inflammation and poor cognition in children. Nutr Rev 2016; 74:374-86. [PMID: 27142301 DOI: 10.1093/nutrit/nuw008] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The intestinal microbiota undergoes active remodeling in the first 6 to 18 months of life, during which time the characteristics of the adult microbiota are developed. This process is strongly influenced by the early diet and enteric pathogens. Enteric infections and malnutrition early in life may favor microbiota dysbiosis and small intestinal bacterial overgrowth, resulting in intestinal barrier dysfunction and translocation of intestinal bacterial products, ultimately leading to low-grade, chronic, subclinical systemic inflammation. The leaky gut-derived low-grade systemic inflammation may have profound consequences on the gut-liver-brain axis, compromising normal growth, metabolism, and cognitive development. This review examines recent data suggesting that early-life enteric infections that lead to intestinal barrier disruption may shift the intestinal microbiota toward chronic systemic inflammation and subsequent impaired cognitive development.
Collapse
Affiliation(s)
- Reinaldo B Oriá
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA.
| | - Laura E Murray-Kolb
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Rebecca J Scharf
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Laura L Pendergast
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Dennis R Lang
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Glynis L Kolling
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Richard L Guerrant
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
131
|
Gause WC, Maizels RM. Macrobiota - helminths as active participants and partners of the microbiota in host intestinal homeostasis. Curr Opin Microbiol 2016; 32:14-18. [PMID: 27116368 DOI: 10.1016/j.mib.2016.04.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/06/2016] [Indexed: 12/21/2022]
Abstract
Important insights have recently been gained in our understanding of the intricate relationship in the intestinal milieu between the vertebrate host mucosal immune response, commensal bacteria, and helminths. Helminths are metazoan worms (macrobiota) and trigger immune responses that include potent regulatory components capable of controlling harmful inflammation, protecting barrier function and mitigating tissue damage. They can secrete a variety of products that directly affect immune regulatory function but they also have the capacity to influence the composition of microbiota, which can also then impact immune function. Conversely, changes in microbiota can affect susceptibility to helminth infection, indicating that crosstalk between these two disparate groups of endobiota can play an essential role in host intestinal immune function and homeostasis.
Collapse
Affiliation(s)
- William C Gause
- RBHS Institute for Infectious and Inflammatory Diseases, Center for Immunity and Inflammation, New Jersey Medical School, Rutgers - The State University of New Jersey, Newark, NJ, USA.
| | - Rick M Maizels
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunology and Inflammation, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
132
|
Giacomin P, Croese J, Krause L, Loukas A, Cantacessi C. Suppression of inflammation by helminths: a role for the gut microbiota? Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0296. [PMID: 26150662 PMCID: PMC4528494 DOI: 10.1098/rstb.2014.0296] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Multiple recent investigations have highlighted the promise of helminth-based therapies for the treatment of inflammatory disorders of the intestinal tract of humans, including inflammatory bowel disease and coeliac disease. However, the mechanisms by which helminths regulate immune responses, leading to the amelioration of symptoms of chronic inflammation are unknown. Given the pivotal roles of the intestinal microbiota in the pathogenesis of these disorders, it has been hypothesized that helminth-induced modifications of the gut commensal flora may be responsible for the therapeutic properties of gastrointestinal parasites. In this article, we review recent progress in the elucidation of host-parasite-microbiota interactions in both animal models of chronic inflammation and humans, and provide a working hypothesis of the role of the gut microbiota in helminth-induced suppression of inflammation.
Collapse
Affiliation(s)
- Paul Giacomin
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield 4878, Australia
| | - John Croese
- Department of Gastroenterology and Hepatology, The Prince Charles Hospital, Brisbane 4007, Australia
| | - Lutz Krause
- Translational Research Institute, University of Queensland Diamantina Institute, Woolloongabba, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield 4878, Australia
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| |
Collapse
|
133
|
Kreisinger J, Bastien G, Hauffe HC, Marchesi J, Perkins SE. Interactions between multiple helminths and the gut microbiota in wild rodents. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0295. [PMID: 26150661 PMCID: PMC4528493 DOI: 10.1098/rstb.2014.0295] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The gut microbiota is vital to host health and, as such, it is important to elucidate the mechanisms altering its composition and diversity. Intestinal helminths are host immunomodulators and have evolved both temporally and spatially in close association with the gut microbiota, resulting in potential mechanistic interplay. Host–helminth and host–microbiota interactions are comparatively well-examined, unlike microbiota–helminth relationships, which typically focus on experimental infection with a single helminth species in laboratory animals. Here, in addition to a review of the literature on helminth–microbiota interactions, we examined empirically the association between microbiota diversity and composition and natural infection of multiple helminth species in wild mice (Apodemus flavicollis), using 16S rRNA gene catalogues (metataxonomics). In general, helminth presence is linked with high microbiota diversity, which may confer health benefits to the host. Within our wild rodent system variation in the composition and abundance of gut microbial taxa associated with helminths was specific to each helminth species and occurred both up- and downstream of a given helminth's niche (gut position). The most pronounced helminth–microbiota association was between the presence of tapeworms in the small intestine and increased S24–7 (Bacteroidetes) family in the stomach. Helminths clearly have the potential to alter gut homeostasis. Free-living rodents with a diverse helminth community offer a useful model system that enables both correlative (this study) and manipulative inference to elucidate helminth–microbiota interactions.
Collapse
Affiliation(s)
- Jakub Kreisinger
- Department of Biodiversity and Molecular Ecology, Centre for Research and Innovation, Fondazione Edmund Mach, Via E. Mach 1, 38010 S. Michele all'Adige, TN, Italy
| | - Géraldine Bastien
- Department of Biodiversity and Molecular Ecology, Centre for Research and Innovation, Fondazione Edmund Mach, Via E. Mach 1, 38010 S. Michele all'Adige, TN, Italy
| | - Heidi C Hauffe
- Department of Biodiversity and Molecular Ecology, Centre for Research and Innovation, Fondazione Edmund Mach, Via E. Mach 1, 38010 S. Michele all'Adige, TN, Italy
| | - Julian Marchesi
- Centre for Digestive and Gut Health, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Sarah E Perkins
- Department of Biodiversity and Molecular Ecology, Centre for Research and Innovation, Fondazione Edmund Mach, Via E. Mach 1, 38010 S. Michele all'Adige, TN, Italy School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| |
Collapse
|
134
|
Microbiome-Epigenome Interactions and the Environmental Origins of Inflammatory Bowel Diseases. J Pediatr Gastroenterol Nutr 2016; 62:208-19. [PMID: 26308318 PMCID: PMC4724338 DOI: 10.1097/mpg.0000000000000950] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The incidence of pediatric inflammatory bowel disease (IBD), which includes Crohn disease and ulcerative colitis, has risen alarmingly in the Western and developing world in recent decades. Epidemiologic (including monozygotic twin and migrant) studies highlight the substantial role of environment and nutrition in IBD etiology. Here we review the literature supporting the developmental and environmental origins hypothesis of IBD. We also provide a detailed exploration of how the human microbiome and epigenome (primarily through DNA methylation) may be important elements in the developmental origins of IBD in both children and adults.
Collapse
|
135
|
Reynolds LA, Finlay BB, Maizels RM. Cohabitation in the Intestine: Interactions among Helminth Parasites, Bacterial Microbiota, and Host Immunity. THE JOURNAL OF IMMUNOLOGY 2016; 195:4059-66. [PMID: 26477048 DOI: 10.4049/jimmunol.1501432] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Both intestinal helminth parasites and certain bacterial microbiota species have been credited with strong immunomodulatory effects. Recent studies reported that the presence of helminth infection alters the composition of the bacterial intestinal microbiota and, conversely, that the presence and composition of the bacterial microbiota affect helminth colonization and persistence within mammalian hosts. This article reviews recent findings on these reciprocal relationships, in both human populations and mouse models, at the level of potential mechanistic pathways and the implications these bear for immunomodulatory effects on allergic and autoimmune disorders. Understanding the multidirectional complex interactions among intestinal microbes, helminth parasites, and the host immune system allows for a more holistic approach when using probiotics, prebiotics, synbiotics, antibiotics, and anthelmintics, as well as when designing treatments for autoimmune and allergic conditions.
Collapse
Affiliation(s)
- Lisa A Reynolds
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada; and
| | - Rick M Maizels
- Centre for Immunity, Infection, and Evolution, Institute of Immunology and Infection Research, Ashworth Laboratories, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| |
Collapse
|
136
|
Izzi JM, Beck SE, Adams RJ, Metcalf Pate KA, Hutchinson EK. Serum Cobalamin (Vitamin B12) Concentrations in Rhesus Macaques (Macaca mulatta) and Pigtailed Macaques (Macaca nemestrina) with Chronic Idiopathic Diarrhea. Comp Med 2016; 66:324-332. [PMID: 27538863 PMCID: PMC4983174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 10/16/2015] [Accepted: 12/06/2015] [Indexed: 06/06/2023]
Abstract
Chronic diarrhea poses a significant threat to the health of NHP research colonies, and its primary etiology remains unclear. In macaques, the clinical presentation of intractable diarrhea and weight loss that are accompanied by inflammatory infiltrates within the gastrointestinal tract closely resembles inflammatory bowel disease of humans, dogs, and cats, in which low serum and tissue cobalamin (vitamin B12) levels are due to intestinal malabsorption. We therefore hypothesized that macaques with chronic idiopathic diarrhea (CID) have lower serum cobalamin concentrations than do healthy macaques. Here we measured serum cobalamin concentrations in both rhesus and pigtailed macaques with CID and compared them with those of healthy controls. Serum cobalamin levels were 2.5-fold lower in pigtailed macaques with CID than control animals but did not differ between rhesus macaques with CID and their controls. This finding supports the use of serum cobalamin concentration as an adjunct diagnostic tool in pigtailed macaques that present with clinical symptoms of chronic gastrointestinal disease. This use of serum vitamin B12 levels has implications for the future use of parenteral cobalamin supplementation to improve clinical outcomes in this species.
Collapse
Affiliation(s)
- Jessica M Izzi
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Division of Veterinary Resources, National Institutes of Health, Bethesda, Maryland, USA.
| | - Sarah E Beck
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Robert J Adams
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kelly A Metcalf Pate
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eric K Hutchinson
- Division of Veterinary Resources, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
137
|
Abstract
Autoimmune and chronic inflammatory organic diseases represent a "postindustrial revolution epidemics," and their frequency has increased dramatically in the last century. Today, it is assumed that the increase in hygiene standards reduced the interactions with helminth parasites that coevolved with the immune system and are crucial for its proper functioning. Several helminths have been proposed and tested in the search of the ideal therapeutic. In this review, the authors summarize the translational and clinical studies and review the caveats and possible solutions for the optimization of helminth therapies.
Collapse
Affiliation(s)
- Irina Leonardi
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zurich, Switzerland
| | - Isabelle Frey
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zurich, Switzerland
| | - Gerhard Rogler
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zurich, Switzerland; Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
138
|
Loke P, Lim YAL. Helminths and the microbiota: parts of the hygiene hypothesis. Parasite Immunol 2015; 37:314-23. [PMID: 25869420 DOI: 10.1111/pim.12193] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/18/2015] [Indexed: 12/12/2022]
Abstract
In modern societies, diseases that are driven by dysregulated immune responses are increasing at an alarming pace, such as inflammatory bowel diseases and diabetes. There is an urgent need to understand these epidemiological trends, which are likely to be driven by the changing environment of the last few decades. There are complex interactions between human genetic factors and this changing environment that is leading to the increasing prevalence of metabolic and inflammatory diseases. Alterations to human gut bacterial communities (the microbiota) and lowered prevalence of helminth infections are potential environmental factors contributing to immune dysregulation. Helminths have co-evolved with the gut microbiota and their mammalian hosts. This three-way interaction is beginning to be characterized, and the knowledge gained may enable the design of new therapeutic strategies to treat metabolic and inflammatory diseases. However, these complex interactions need to be carefully investigated in the context of host genetic backgrounds to identify optimal treatment strategies. The complex nature of these interactions raises the possibility that only with highly personalized treatment, with knowledge of individual genetic and microbiota communities, will therapeutic interventions be successful for a majority of the individuals suffering from these complex diseases of immune dysregulation.
Collapse
Affiliation(s)
- P Loke
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | | |
Collapse
|
139
|
Zaiss MM, Rapin A, Lebon L, Dubey LK, Mosconi I, Sarter K, Piersigilli A, Menin L, Walker AW, Rougemont J, Paerewijck O, Geldhof P, McCoy KD, Macpherson AJ, Croese J, Giacomin PR, Loukas A, Junt T, Marsland BJ, Harris NL. The Intestinal Microbiota Contributes to the Ability of Helminths to Modulate Allergic Inflammation. Immunity 2015; 43:998-1010. [PMID: 26522986 PMCID: PMC4658337 DOI: 10.1016/j.immuni.2015.09.012] [Citation(s) in RCA: 321] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 07/12/2015] [Accepted: 09/28/2015] [Indexed: 12/14/2022]
Abstract
Intestinal helminths are potent regulators of their host’s immune system and can ameliorate inflammatory diseases such as allergic asthma. In the present study we have assessed whether this anti-inflammatory activity was purely intrinsic to helminths, or whether it also involved crosstalk with the local microbiota. We report that chronic infection with the murine helminth Heligmosomoides polygyrus bakeri (Hpb) altered the intestinal habitat, allowing increased short chain fatty acid (SCFA) production. Transfer of the Hpb-modified microbiota alone was sufficient to mediate protection against allergic asthma. The helminth-induced anti-inflammatory cytokine secretion and regulatory T cell suppressor activity that mediated the protection required the G protein-coupled receptor (GPR)-41. A similar alteration in the metabolic potential of intestinal bacterial communities was observed with diverse parasitic and host species, suggesting that this represents an evolutionary conserved mechanism of host-microbe-helminth interactions. The microbiota contributes to helminth-induced modulation of allergic asthma Cecal microbial communities are altered in helminth-infected mice Helminth infection increases microbial-derived short chain fatty acids GPR41 mediates helminth-induced Treg cell suppressor function
Collapse
Affiliation(s)
- Mario M Zaiss
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Alexis Rapin
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Luc Lebon
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Lalit Kumar Dubey
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Ilaria Mosconi
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Kerstin Sarter
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland
| | - Alessandra Piersigilli
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland; Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern 3012, Switzerland
| | - Laure Menin
- Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Alan W Walker
- Pathogen Genomics Group, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, UK; Microbiology Group, Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen AB21 9SB, UK
| | - Jacques Rougemont
- Bioinformatics and Biostatistics Core Facility, École Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Oonagh Paerewijck
- Department of Virology, Parasitology and Immunology, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Peter Geldhof
- Department of Virology, Parasitology and Immunology, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Kathleen D McCoy
- Maurice Müller Laboratories (DKF), University Hospital of Bern, Bern 3010, Switzerland
| | - Andrew J Macpherson
- Maurice Müller Laboratories (DKF), University Hospital of Bern, Bern 3010, Switzerland
| | - John Croese
- Department of Gastroenterology and Hepatology, The Prince Charles Hospital, Chermside, Brisbane, QLD 4032, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD 4870, Australia
| | - Paul R Giacomin
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD 4870, Australia
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD 4870, Australia
| | | | - Benjamin J Marsland
- Faculty of Biology and Medicine, University of Lausanne, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne 1011, Switzerland
| | - Nicola L Harris
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland.
| |
Collapse
|
140
|
Giacomin P, Zakrzewski M, Croese J, Su X, Sotillo J, McCann L, Navarro S, Mitreva M, Krause L, Loukas A, Cantacessi C. Experimental hookworm infection and escalating gluten challenges are associated with increased microbial richness in celiac subjects. Sci Rep 2015; 5:13797. [PMID: 26381211 PMCID: PMC4585380 DOI: 10.1038/srep13797] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/04/2015] [Indexed: 02/08/2023] Open
Abstract
The intestinal microbiota plays a critical role in the development of the immune system. Recent investigations have highlighted the potential of helminth therapy for treating a range of inflammatory disorders, including celiac disease (CeD); however, the mechanisms by which helminths modulate the immune response of the human host and ameliorate CeD pathology are unknown. In this study, we investigated the potential role of alterations in the human gut microbiota in helminth-mediated suppression of an inflammatory disease. We assessed the qualitative and quantitative changes in the microbiota of human volunteers with CeD prior to and following infection with human hookworms, and following challenge with escalating doses of dietary gluten. Experimental hookworm infection of the trial subjects resulted in maintenance of the composition of the intestinal flora, even after a moderate gluten challenge. Notably, we observed a significant increase in microbial species richness over the course of the trial, which could represent a potential mechanism by which hookworms can regulate gluten-induced inflammation and maintain intestinal immune homeostasis.
Collapse
Affiliation(s)
- Paul Giacomin
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Martha Zakrzewski
- Bioinformatics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - John Croese
- Prince Charles Hospital, Brisbane, QLD, Australia
| | - Xiaopei Su
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Javier Sotillo
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Leisa McCann
- Prince Charles Hospital, Brisbane, QLD, Australia
| | - Severine Navarro
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Makedonka Mitreva
- The Genome Institute, and.,Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lutz Krause
- Bioinformatics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Cinzia Cantacessi
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.,Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
141
|
Study of the gastrointestinal parasitic fauna of captive non-human primates (Macaca fascicularis). Parasitol Res 2015; 115:307-12. [PMID: 26374536 DOI: 10.1007/s00436-015-4748-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/08/2015] [Indexed: 12/24/2022]
Abstract
The aim of this study was to examine helminths and protozoans in cynomolgus macaques (Macaca fascicularis) imported from registered breeding facilities in China and their relation to health risks for non-human primate handlers in biomedical research centers and in breeding facilities. Fresh fecal samples were collected from a total of 443 M. fascicularis and analyzed by copromicroscopical analysis, immunoenzymatic, or molecular assays. As to helminths, whose eggs were shed in 2.03% of the samples, Trichuris and Oesophagostomum were the only two taxa found, with low prevalence and low eggs per gram (EPG) values. Protozoans were more frequently detected (87.40%), with Entamoeba coli (85.19%) and Endolimax nana (79.26%) as the most prevalent species shed. Other parasites found by fecal smear examination were uninucleated-cyst-producing Entamoebas (78.52%), Iodamoeba bütschlii (42.96%), and Chilomastix mesnili (24.44%), while cysts of Balantidium coli (22.2%) were only observed by sedimentation. No coproantigens of Giardia duodenalis, Cryptosporidium spp., and Entamoeba histolytica complex were detected. Blastocystis sp. infection was noticed in 87.63% of macaques by PCR. These cynomolgus monkeys were infected with many subtypes (ST1, ST2, ST3, ST5, and ST7), where the predominant Blastocystis sp. subtypes were ST2 (77.5%), followed by ST1 (63.5%). Data collected confirmed the presence of potentially zoonotic parasites and a high parasite diversity, suggesting the need for appropriate and sensitive techniques to adequately control them and related health risks for handlers of non-human primates in biomedical research centers and in breeding facilities.
Collapse
|
142
|
Kay GL, Millard A, Sergeant MJ, Midzi N, Gwisai R, Mduluza T, Ivens A, Nausch N, Mutapi F, Pallen M. Differences in the Faecal Microbiome in Schistosoma haematobium Infected Children vs. Uninfected Children. PLoS Negl Trop Dis 2015; 9:e0003861. [PMID: 26114287 PMCID: PMC4482744 DOI: 10.1371/journal.pntd.0003861] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 05/31/2015] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Several infectious diseases and therapeutic interventions cause gut microbe dysbiosis and associated pathology. We characterised the gut microbiome of children exposed to the helminth Schistosoma haematobium pre- and post-treatment with the drug praziquantel (PZQ), with the aim to compare the gut microbiome structure (abundance and diversity) in schistosome infected vs. uninfected children. METHODS Stool DNA from 139 children aged six months to 13 years old; with S. haematobium infection prevalence of 27.34% was extracted at baseline. 12 weeks following antihelminthic treatment with praziqunatel, stool DNA was collected from 62 of the 139 children. The 16S rRNA genes were sequenced from the baseline and post-treatment samples and the sequence data, clustered into operational taxonomic units (OTUs). The OTU data were analysed using multivariate analyses and paired T-test. RESULTS Pre-treatment, the most abundant phyla were Bacteroidetes, followed by Firmicutes and Proteobacteria respectively. The relative abundance of taxa among bacterial classes showed limited variation by age group or sex and the bacterial communities had similar overall compositions. Although there were no overall differences in the microbiome structure across the whole age range, the abundance of 21 OTUs varied significantly with age (FDR<0.05). Some OTUs including Veillonella, Streptococcus, Bacteroides and Helicobacter were more abundant in children ≤ 1 year old compared to older children. Furthermore, the gut microbiome differed in schistosome infected vs. uninfected children with 27 OTU occurring in infected but not uninfected children, for 5 of these all Prevotella, the difference was statistically significant (p <0.05) with FDR <0.05. PZQ treatment did not alter the microbiome structure in infected or uninfected children from that observed at baseline. CONCLUSIONS There are significant differences in the gut microbiome structure of infected vs. uninfected children and the differences were refractory to PZQ treatment.
Collapse
Affiliation(s)
- Gemma Louise Kay
- Microbiology and Infection Unit, Division of Translational and Systems Medicine, Warwick Medical School, University of Warwick, Gibbet Hill Campus, Coventry, West Midlands, United Kingdom
| | - Andrew Millard
- Microbiology and Infection Unit, Division of Translational and Systems Medicine, Warwick Medical School, University of Warwick, Gibbet Hill Campus, Coventry, West Midlands, United Kingdom
| | - Martin J. Sergeant
- Microbiology and Infection Unit, Division of Translational and Systems Medicine, Warwick Medical School, University of Warwick, Gibbet Hill Campus, Coventry, West Midlands, United Kingdom
| | - Nicholas Midzi
- National Institute of Health Research, Causeway, Harare, Zimbabwe
| | | | - Takafira Mduluza
- Department of Biochemistry, University of Zimbabwe, Mount Pleasant, Harare, Zimbabwe
| | - Alasdair Ivens
- Insitute of Immunology and Infection Research, Centre for Immunology, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, King’s Buildings, Edinburgh, United Kingdom
| | - Norman Nausch
- Insitute of Immunology and Infection Research, Centre for Immunology, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, King’s Buildings, Edinburgh, United Kingdom
| | - Francisca Mutapi
- Insitute of Immunology and Infection Research, Centre for Immunology, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, King’s Buildings, Edinburgh, United Kingdom
| | - Mark Pallen
- Microbiology and Infection Unit, Division of Translational and Systems Medicine, Warwick Medical School, University of Warwick, Gibbet Hill Campus, Coventry, West Midlands, United Kingdom
| |
Collapse
|
143
|
McKay DM. Not all parasites are protective. Parasite Immunol 2015; 37:324-32. [DOI: 10.1111/pim.12160] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 11/09/2014] [Indexed: 12/11/2022]
Affiliation(s)
- Derek M. McKay
- Department of Physiology and Pharmacology; Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases; Gastrointestinal Research Group and Inflammation Research Network; Cumming School of Medicine, University of Calgary; Calgary AB Canada
| |
Collapse
|
144
|
McKenney EA, Williamson L, Yoder AD, Rawls JF, Bilbo SD, Parker W. Alteration of the rat cecal microbiome during colonization with the helminth Hymenolepis diminuta. Gut Microbes 2015; 6:182-93. [PMID: 25942385 PMCID: PMC4615828 DOI: 10.1080/19490976.2015.1047128] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The microbiome is now widely recognized as being important in health and disease, and makes up a substantial subset of the biome within the ecosystem of the vertebrate body. At the same time, multicellular, eukaryotic organisms such as helminths are being recognized as an important component of the biome that shaped the evolution of our genes. The absence of these macroscopic organisms during the early development and life of humans in Western culture probably leads to a wide range of human immunological diseases. However, the interaction between the microbiome and macroscopic components of the biome remains poorly characterized. In this study, the microbiome of the cecum in rats colonized for 2 generations with the small intestinal helminth Hymenolepis diminuta was evaluated. The introduction of this benign helminth, which is of considerable therapeutic interest, led to several changes in the cecal microbiome. Most of the changes were within the Firmicutes phylum, involved about 20% of the total bacteria, and generally entailed a shift from Bacilli to Clostridia species in the presence of the helminth. The results point toward ecological relationships between various components of the biome, with the observed shifts in the microbiome suggesting potential mechanisms by which this helminth might exert therapeutic effects.
Collapse
Affiliation(s)
| | | | - Anne D Yoder
- Departments of Biology; Duke University; Durham, NC, USA,Evolutionary Anthropology; Duke University; Durham, NC, USA
| | - John F Rawls
- Molecular Genetics and Microbiology; Duke University; Durham, NC, USA
| | - Staci D Bilbo
- Psychology & Neuroscience; Duke University; Durham, NC, USA
| | - William Parker
- Department of Surgery; Duke University Medical Center; Durham, NC, USA,Correspondence to: William Parker;
| |
Collapse
|
145
|
Holm JB, Sorobetea D, Kiilerich P, Ramayo-Caldas Y, Estellé J, Ma T, Madsen L, Kristiansen K, Svensson-Frej M. Chronic Trichuris muris Infection Decreases Diversity of the Intestinal Microbiota and Concomitantly Increases the Abundance of Lactobacilli. PLoS One 2015; 10:e0125495. [PMID: 25942314 PMCID: PMC4420551 DOI: 10.1371/journal.pone.0125495] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/13/2015] [Indexed: 12/26/2022] Open
Abstract
The intestinal microbiota is vital for shaping the local intestinal environment as well as host immunity and metabolism. At the same time, epidemiological and experimental evidence suggest an important role for parasitic worm infections in maintaining the inflammatory and regulatory balance of the immune system. In line with this, the prevalence of persistent worm infections is inversely correlated with the incidence of immune-associated diseases, prompting the use of controlled parasite infections for therapeutic purposes. Despite this, the impact of parasite infection on the intestinal microbiota, as well as potential downstream effects on the immune system, remain largely unknown. We have assessed the influence of chronic infection with the large-intestinal nematode Trichuris muris, a close relative of the human pathogen Trichuris trichiura, on the composition of the murine intestinal microbiota by 16S ribosomal-RNA gene-based sequencing. Our results demonstrate that persistent T. muris infection dramatically affects the large-intestinal microbiota, most notably with a drop in the diversity of bacterial communities, as well as a marked increase in the relative abundance of the Lactobacillus genus. In parallel, chronic T. muris infection resulted in a significant shift in the balance between regulatory and inflammatory T cells in the intestinal adaptive immune system, in favour of inflammatory cells. Together, these data demonstrate that chronic parasite infection strongly influences the intestinal microbiota and the adaptive immune system. Our results illustrate the complex interactions between these factors in the intestinal tract, and contribute to furthering the understanding of this interplay, which is of crucial importance considering that 500 million people globally are suffering from these infections and their potential use for therapeutic purposes.
Collapse
Affiliation(s)
- Jacob Bak Holm
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Sorobetea
- Immunology Section, Department of Experimental Medical Sciences, Medical Faculty, Lund University, Lund, Sweden
| | - Pia Kiilerich
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Jordi Estellé
- INRA, UMR1313 Génétique Animale et Biologie Intégrative (GABI), Jouy-en-Josas, France
| | - Tao Ma
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Lise Madsen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark; National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Marcus Svensson-Frej
- Immunology Section, Department of Experimental Medical Sciences, Medical Faculty, Lund University, Lund, Sweden
| |
Collapse
|
146
|
Houlden A, Hayes KS, Bancroft AJ, Worthington JJ, Wang P, Grencis RK, Roberts IS. Chronic Trichuris muris Infection in C57BL/6 Mice Causes Significant Changes in Host Microbiota and Metabolome: Effects Reversed by Pathogen Clearance. PLoS One 2015; 10:e0125945. [PMID: 25938477 PMCID: PMC4418675 DOI: 10.1371/journal.pone.0125945] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/25/2015] [Indexed: 12/26/2022] Open
Abstract
Trichuris species are a globally important and prevalent group of intestinal helminth parasites, in which Trichuris muris (mouse whipworm) is an ideal model for this disease. This paper describes the first ever highly controlled and comprehensive investigation into the effects of T. muris infection on the faecal microbiota of mice and the effects on the microbiota following successful clearance of the infection. Communities were profiled using DGGE, 454 pyrosequencing, and metabolomics. Changes in microbial composition occurred between 14 and 28 days post infection, resulting in significant changes in α and β- diversity. This impact was dominated by a reduction in the diversity and abundance of Bacteroidetes, specifically Prevotella and Parabacteroides. Metabolomic analysis of stool samples of infected mice at day 41 showed significant differences to uninfected controls with a significant increase in the levels of a number of essential amino acids and a reduction in breakdown of dietary plant derived carbohydrates. The significant reduction in weight gain by infected mice probably reflects these metabolic changes and the incomplete digestion of dietary polysaccharides. Following clearance of infection the intestinal microbiota underwent additional changes gradually transitioning by day 91 towards a microbiota of an uninfected animal. These data indicate that the changes in microbiota as a consequence of infection were transitory requiring the presence of the pathogen for maintenance. Interestingly this was not observed for all of the key immune cell populations associated with chronic T. muris infection. This reflects the highly regulated chronic response and potential lasting immunological consequences of dysbiosis in the microbiota. Thus infection of T. muris causes a significant and substantial impact on intestinal microbiota and digestive function of mice with affects in long term immune regulation.
Collapse
Affiliation(s)
- Ashley Houlden
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Kelly S. Hayes
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Allison J. Bancroft
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - John J. Worthington
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Ping Wang
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Richard K. Grencis
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
- * E-mail: (RKG); (ISR)
| | - Ian S. Roberts
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
- * E-mail: (RKG); (ISR)
| |
Collapse
|
147
|
Grencis RK. Immunity to Helminths: Resistance, Regulation, and Susceptibility to Gastrointestinal Nematodes. Annu Rev Immunol 2015; 33:201-25. [DOI: 10.1146/annurev-immunol-032713-120218] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Richard K. Grencis
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| |
Collapse
|
148
|
Chachage M, Geldmacher C. Immune system modulation by helminth infections: potential impact on HIV transmission and disease progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 828:131-49. [PMID: 25253030 DOI: 10.1007/978-1-4939-1489-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Affiliation(s)
- Mkunde Chachage
- Department of Cellular Immunology, National Institute for Medical Research-Mbeya Medical Research Centre (NIMR-MMRC), Hospital Hill road, Mbeya, Tanzania,
| | | |
Collapse
|
149
|
Grencis RK, Humphreys NE, Bancroft AJ. Immunity to gastrointestinal nematodes: mechanisms and myths. Immunol Rev 2015; 260:183-205. [PMID: 24942690 PMCID: PMC4141702 DOI: 10.1111/imr.12188] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immune responses to gastrointestinal nematodes have been studied extensively for over 80 years and intensively investigated over the last 30–40 years. The use of laboratory models has led to the discovery of new mechanisms of protective immunity and made major contributions to our fundamental understanding of both innate and adaptive responses. In addition to host protection, it is clear that immunoregulatory processes are common in infected individuals and resistance often operates alongside modulation of immunity. This review aims to discuss the recent discoveries in both host protection and immunoregulation against gastrointestinal nematodes, placing the data in context of the specific life cycles imposed by the different parasites studied and the future challenges of considering the mucosal/immune axis to encompass host, parasite, and microbiome in its widest sense.
Collapse
|
150
|
Ferretti CJ, Hollander E. The Role of Inflammation in Autism Spectrum Disorder. CURRENT TOPICS IN NEUROTOXICITY 2015. [DOI: 10.1007/978-3-319-13602-8_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|