51
|
Mejia R, Damania A, Jeun R, Bryan PE, Vargas P, Juarez M, Cajal PS, Nasser J, Krolewiecki A, Lefoulon E, Long C, Drake E, Cimino RO, Slatko B. Impact of intestinal parasites on microbiota and cobalamin gene sequences: a pilot study. Parasit Vectors 2020; 13:200. [PMID: 32306993 PMCID: PMC7168842 DOI: 10.1186/s13071-020-04073-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/10/2020] [Indexed: 01/10/2023] Open
Abstract
Background Approximately 30% of children worldwide are infected with gastrointestinal parasites. Depending on the species, parasites can disrupt intestinal bacterial microbiota affecting essential vitamin biosynthesis. Methods Stool samples were collected from 37 asymptomatic children from a previous cross-sectional Argentinian study. A multi-parallel real-time quantitative PCR was implemented for Ascaris lumbricoides, Ancylostoma duodenale, Necator americanus, Strongyloides stercoralis, Trichuris trichiura, Cryptosporidium spp., Entamoeba histolytica and Giardia duodenalis. In addition, whole-genome sequencing analysis was conducted for bacterial microbiota on all samples and analyzed using Livermore Metagenomic Analysis Toolkit and DIAMOND software. Separate analyses were carried out for uninfected, Giardia-only, Giardia + helminth co-infections, and helminth-only groups. Results For Giardia-only infected children compared to uninfected children, DNA sequencing data showed a decrease in microbiota biodiversity that correlated with increasing Giardia burden and was statistically significant using Shannonʼs alpha diversity (Giardia-only > 1 fg/µl 2.346; non-infected group 3.253, P = 0.0317). An increase in diversity was observed for helminth-only infections with a decrease in diversity for Giardia + helminth co-infections (P = 0.00178). In Giardia-only infections, microbiome taxonomy changed from Firmicutes towards increasing proportions of Prevotella, with the degree of change related to the intensity of infection compared to uninfected (P = 0.0317). The abundance of Prevotella bacteria was decreased in the helminths-only group but increased for Giardia + helminth co-infections (P = 0.0262). Metagenomic analysis determined cobalamin synthesis was decreased in the Giardia > 1 fg/µl group compared to both the Giardia < 1 fg/µl and the uninfected group (P = 0.0369). Giardia + helminth group also had a decrease in cobalamin CbiM genes from helminth-only infections (P = 0.000754). Conclusion The study results may provide evidence for an effect of parasitic infections enabling the permissive growth of anaerobic bacteria such as Prevotella, suggesting an altered capacity of vitamin B12 (cobalamin) biosynthesis and potential impact on growth and development in children .
Collapse
Affiliation(s)
- Rojelio Mejia
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA. .,Universidad Nacional de Salta, Salta, Argentina.
| | - Ashish Damania
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Rebecca Jeun
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Patricia E Bryan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | | | | | | | | | | | | | - Evan Drake
- New England Biolabs, Inc, Ipswich, MA, USA
| | | | | |
Collapse
|
52
|
The Chymase Mouse Mast Cell Protease-4 Regulates Intestinal Cytokine Expression in Mature Adult Mice Infected with Giardia intestinalis. Cells 2020; 9:cells9040925. [PMID: 32283818 PMCID: PMC7226739 DOI: 10.3390/cells9040925] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022] Open
Abstract
Mast cells have been shown to affect the control of infections with the protozoan parasite Giardia intestinalis. Recently, we demonstrated that Giardia excretory-secretory proteins inhibited the activity of the connective tissue mast cell-specific protease chymase. To study the potential role of the chymase mouse mast cell protease (mMCP)-4 during infections with Giardia, mMCP-4+/+ and mMCP-4−/− littermate mice were gavage-infected with G. intestinalis trophozoites of the human assemblage B isolate GS. No significant changes in weight gain was observed in infected young (≈10 weeks old) mMCP-4−/− and mMCP-4+/+ littermate mice. In contrast, infections of mature adult mice (>18 weeks old) caused significant weight loss as compared to uninfected control mice. We detected a more rapid weight loss in mMCP-4−/− mice as compared to littermate mMCP-4+/+ mice. Submucosal mast cell and granulocyte counts in jejunum increased in the infected adult mMCP-4−/− and mMCP-4+/+ mice. This increase was correlated with an augmented intestinal trypsin-like and chymotrypsin-like activity, but the myeloperoxidase activity was constant. Infected mice showed a significantly lower intestinal neutrophil elastase (NE) activity, and in vitro, soluble Giardia proteins inhibited human recombinant NE. Serum levels of IL-6 were significantly increased eight and 13 days post infection (dpi), while intestinal IL-6 levels showed a trend to significant increase 8 dpi. Strikingly, the lack of mMCP-4 resulted in significantly less intestinal transcriptional upregulation of IL-6, TNF-α, IL-25, CXCL2, IL-2, IL-4, IL-5, and IL-10 in the Giardia-infected mature adult mice, suggesting that chymase may play a regulatory role in intestinal cytokine responses.
Collapse
|
53
|
Buret AG. Acceptance of the 2019 Stoll-Stunkard Memorial Lectureship Award: The Study of Host-Parasite Interactions to Better Understand Fundamental Host Physiology: The Model of Giardiasis. J Parasitol 2020. [DOI: 10.1645/19-134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- André G. Buret
- Department of Biological Sciences, Host-Parasite Interactions Program, Inflammation Research Network, University of Calgary, 2500 University Drive N.W., Calgary (Alberta), T2N 1N4, Canada
| |
Collapse
|
54
|
Fredensborg BL, Fossdal í Kálvalíð I, Johannesen TB, Stensvold CR, Nielsen HV, Kapel CMO. Parasites modulate the gut-microbiome in insects: A proof-of-concept study. PLoS One 2020; 15:e0227561. [PMID: 31935259 PMCID: PMC6959588 DOI: 10.1371/journal.pone.0227561] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/21/2019] [Indexed: 01/18/2023] Open
Abstract
Host-parasite interactions may be modulated by host- or parasite-associated microbes, but the role of these are often overlooked. Particularly for parasites with intestinal stages (either larval or adult), the host gut microbiome may play a key role for parasite establishment; moreover, the microbiome may change in response to invading parasites. Hypothesis testing at the organismal level may be hampered, particularly in mammalian definitive hosts, by ethical, logistical, and economical restrictions. Thus, invertebrates naturally serving as intermediate hosts to parasites with complex life cycles may inform the development of mammalian models as an early-stage host-parasite model. In addition, several important pathogens are vectored by insects, and insect gut microbiome-pathogen interactions may provide essential base-line knowledge, which may be used to control vectorborne pathogens. Here, we used the grain beetle, Tenebrio molitor, a host of the tapeworm Hymenolepis diminuta, to explore interactions between infection status and resident gut microbiota at two pre-determined time points (day two and seven) post infection. Using 16S/18S microbial profiling, we measured key parameters of the composition, relative abundance, and diversity of the host gut bacteriome and mycobiome. In addition, we quantified the systemic beetle immune response to infection by Phenoloxidase activity and hemocyte abundance. We found significant changes in the gut bacteriome and mycobiome in relation to infection status and beetle age. Thus, the relative abundance of Proteobacteria was significantly higher in the gut of infected beetles and driven mostly by an increased abundance of Acinetobacter. In addition, the mycobiome was less abundant in infected beetles but maintained higher Shannon diversity in infected compared with non-infected beetles. Beetles treated with a broad-spectrum antibiotic (Tetracycline) exhibited significantly reduced parasite establishment compared with the untreated control group, indicating that the host microbiome may greatly influence hatching of eggs and subsequent establishment of H. diminuta larvae. Our results suggest that experimental work using invertebrates may provide a platform for explorative studies of host-parasite-microbe interactions and their underlying mechanisms.
Collapse
Affiliation(s)
- Brian L. Fredensborg
- Section for Organismal Biology, Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark
- * E-mail:
| | - Inga Fossdal í Kálvalíð
- Section for Organismal Biology, Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Thor B. Johannesen
- Department of Microbiology and Infection Control, Statens Serum Institut, Copenhagen, Denmark
| | - C. Rune Stensvold
- Department of Microbiology and Infection Control, Statens Serum Institut, Copenhagen, Denmark
| | - Henrik V. Nielsen
- Department of Microbiology and Infection Control, Statens Serum Institut, Copenhagen, Denmark
| | - Christian M. O. Kapel
- Section for Organismal Biology, Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
55
|
RORγt + Treg to Th17 ratios correlate with susceptibility to Giardia infection. Sci Rep 2019; 9:20328. [PMID: 31889073 PMCID: PMC6937251 DOI: 10.1038/s41598-019-56416-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022] Open
Abstract
Infections with Giardia are among the most common causes of food and water-borne diarrheal disease worldwide. Here, we investigated Th17, Treg and IgA responses, and alterations in gut microbiota in two mouse lines with varying susceptibility to Giardia muris infection. Infected BALB/c mice shed significantly more cysts compared with C57BL/6 mice. Impaired control of infection in BALB/c mice was associated with lower Th17 activity and lower IgA levels compared with C57BL/6 mice. The limited metabolic activity, proliferation and cytokine production of Th17 cells in BALB/c mice was associated with higher proportions of intestinal Foxp3+RORγt+ regulatory T cells and BALB/c mice developed increased RORγt+ Treg:Th17 ratios in response to G. muris infection. Furthermore, G. muris colonization led to a significantly reduced evenness in the gut microbial communities of BALB/c mice. Our data indicate that differential susceptibility to Giardia infections may be related to RORγt+ Treg controlling Th17 activity and that changes in the microbiota composition upon Giardia infection partially depend on the host background.
Collapse
|
56
|
Allain T, Buret AG. Pathogenesis and post-infectious complications in giardiasis. ADVANCES IN PARASITOLOGY 2019; 107:173-199. [PMID: 32122529 DOI: 10.1016/bs.apar.2019.12.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Giardia is an important cause of diarrhoea, and results in post-infectious and extra-intestinal complications. This chapter presents a state-of-the art of our understanding of how this parasite may cause such abnormalities, which appear to develop at least in part in Assemblage-dependent manner. Findings from prospective longitudinal cohort studies indicate that Giardia is one of the four most prevalent enteropathogens in early life, and represents a risk factor for stunting at 2 years of age. This may occur independently of diarrheal disease, in strong support of the pathophysiological significance of the intestinal abnormalities induced by this parasite. These include epithelial malabsorption and maldigestion, increased transit, mucus depletion, and disruptions of the commensal microbiota. Giardia increases epithelial permeability and facilitates the invasion of gut bacteria. Loss of intestinal barrier function is at the core of the acute and post-infectious complications associated with this infection. Recent findings demonstrate that the majority of the pathophysiological responses triggered by this parasite can be recapitulated by the effects of its membrane-bound and secreted cysteine proteases.
Collapse
Affiliation(s)
- Thibault Allain
- University of Calgary, Host-Parasite Interactions Program, Inflammation Research Network, Department of Biological Sciences, Calgary, Canada
| | - André G Buret
- University of Calgary, Host-Parasite Interactions Program, Inflammation Research Network, Department of Biological Sciences, Calgary, Canada.
| |
Collapse
|
57
|
Hooks KB, O'Malley MA. Contrasting Strategies: Human Eukaryotic Versus Bacterial Microbiome Research. J Eukaryot Microbiol 2019; 67:279-295. [PMID: 31583780 PMCID: PMC7154641 DOI: 10.1111/jeu.12766] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/11/2019] [Accepted: 09/25/2019] [Indexed: 12/25/2022]
Abstract
Most discussions of human microbiome research have focused on bacterial investigations and findings. Our target is to understand how human eukaryotic microbiome research is developing, its potential distinctiveness, and how problems can be addressed. We start with an overview of the entire eukaryotic microbiome literature (578 papers), show tendencies in the human‐based microbiome literature, and then compare the eukaryotic field to more developed human bacterial microbiome research. We are particularly concerned with problems of interpretation that are already apparent in human bacterial microbiome research (e.g. disease causality, probiotic interventions, evolutionary claims). We show where each field converges and diverges, and what this might mean for progress in human eukaryotic microbiome research. Our analysis then makes constructive suggestions for the future of the field.
Collapse
Affiliation(s)
- Katarzyna B Hooks
- CBiB, University of Bordeaux, Bordeaux, 33076, France.,CNRS/LaBRI, University of Bordeaux, Talence, 33405, France
| | - Maureen A O'Malley
- School of History and Philosophy of Science, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
58
|
Rueckert S, Betts EL, Tsaousis AD. The Symbiotic Spectrum: Where Do the Gregarines Fit? Trends Parasitol 2019; 35:687-694. [DOI: 10.1016/j.pt.2019.06.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 02/06/2023]
|
59
|
Wang C, Li Q, Ren J. Microbiota-Immune Interaction in the Pathogenesis of Gut-Derived Infection. Front Immunol 2019; 10:1873. [PMID: 31456801 PMCID: PMC6698791 DOI: 10.3389/fimmu.2019.01873] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022] Open
Abstract
Gut-derived infection is among the most common complications in patients who underwent severe trauma, serious burn, major surgery, hemorrhagic shock or severe acute pancreatitis (SAP). It could cause sepsis and multiple organ dysfunction syndrome (MODS), which are regarded as a leading cause of mortality in these cases. Gut-derived infection is commonly caused by pathological translocation of intestinal bacteria or endotoxins, resulting from the dysfunction of the gut barrier. In the last decades, the studies regarding to the pathogenesis of gut-derived infection mainly focused on the breakdown of intestinal epithelial tight junction and increased permeability. Limited information is available on the roles of intestinal microbial barrier in the development of gut-derived infection. Recently, advances of next-generation DNA sequencing techniques and its utilization has revolutionized the gut microecology, leading to novel views into the composition of the intestinal microbiota and its connections with multiple diseases. Here, we reviewed the recent progress in the research field of intestinal barrier disruption and gut-derived infection, mainly through the perspectives of the dysbiosis of intestinal microbiota and its interaction with intestinal mucosal immune cells. This review presents novel insights into how the gut microbiota collaborates with mucosal immune cells to involve the development of pathological bacterial translocation. The data might have important implication to better understand the mechanism underlying pathological bacterial translocation, contributing us to develop new strategies for prevention and treatment of gut-derived sepsis.
Collapse
Affiliation(s)
| | - Qiurong Li
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
60
|
Singer SM, Fink MY, Angelova VV. Recent insights into innate and adaptive immune responses to Giardia. ADVANCES IN PARASITOLOGY 2019; 106:171-208. [PMID: 31630758 DOI: 10.1016/bs.apar.2019.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Infection with Giardia produces a wide range of clinical outcomes. Acutely infected patients may have no overt symptoms or suffer from severe cramps, diarrhea, nausea and even urticaria. Recently, post-infectious irritable bowel syndrome and chronic fatigue syndrome have been identified as long-term sequelae of giardiasis. Frequently, recurrent and chronic Giardia infection is considered a major contributor to stunting in children from low and middle income countries. Perhaps the most unusual outcome of infection with Giardia is the apparent reduced risk of developing moderate-to-severe diarrhea due to other enteric infections which has been noted in several recent studies. The goal of understanding immune responses against Giardia is therefore to identify protective mechanisms which could become targets for vaccine development, but also to identify mechanisms whereby infections lead to these other diverse outcomes. Giardia induces a robust adaptive immune response in both humans and animals. It has been known for many years that there is production of large amounts of parasite-specific IgA following infection and that CD4+ T cell responses contribute to this IgA production and control of the infection. In the past decade, there have been advances in our understanding of the non-antibody effector mechanisms used by the host to fight Giardia infections, in particular the importance of the cytokine interleukin (IL)-17 in orchestrating these responses. There have also been major advances in understanding how the innate response to Giardia infection is initiated and how it contributes to the development of adaptive immunity. Finally, there here have been significant increases in our knowledge of how the resident microbial community influences the immune response and how these responses contribute to the development of some of the symptoms of giardiasis. In this article, we will focus on data generated in the last 10 years and how it has advanced our knowledge about this important parasitic disease.
Collapse
Affiliation(s)
- Steven M Singer
- Department of Biology, Georgetown University, Washington, DC, United States.
| | - Marc Y Fink
- Department of Biology, Georgetown University, Washington, DC, United States
| | - Vanessa V Angelova
- Department of Biology, Georgetown University, Washington, DC, United States
| |
Collapse
|
61
|
Thompson RCA, Ash A. Molecular epidemiology of Giardia and Cryptosporidium infections - What's new? INFECTION GENETICS AND EVOLUTION 2019; 75:103951. [PMID: 31279819 DOI: 10.1016/j.meegid.2019.103951] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/30/2019] [Accepted: 07/02/2019] [Indexed: 12/22/2022]
Abstract
New information generated since 2016 from the application of molecular tools to infections with Giardia and Cryptosporidium is critically summarised. In the context of molecular epidemiology, nomenclature, taxonomy, in vitro culture, detection, zoonoses, population genetics and pathogenicity, are covered. Whole genome sequencing has had the greatest impact in the last three years. Future advances will provide a much better understanding of the zoonotic potential of both parasites, their diversity and how this is linked to pathogenesis in different hosts.
Collapse
Affiliation(s)
- R C A Thompson
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia.
| | - A Ash
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia
| |
Collapse
|
62
|
Cryptosporidium parvum-Infected Neonatal Mice Show Gut Microbiota Remodelling Using High-Throughput Sequencing Analysis: Preliminary Results. Acta Parasitol 2019; 64:268-275. [PMID: 30915719 DOI: 10.2478/s11686-019-00044-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/19/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND During the last decade, the scientific community has begun to investigate the composition and role of gut microbiota in normal health and disease. These studies have provided crucial information on the relationship between gut microflora composition and intestinal parasitic infection, and have demonstrated that many enteric pathogen infections are associated with altered gut microflora composition. In this study, we investigated the effects of Cryptosporidium parvum infection (zoonotic protozoan affecting a large range of vertebrates) on both qualitative and quantitative composition of gut microbiota in a CD-1 neonatal mouse model. METHODS 5-day-old neonate mice were experimentally infected with 105Cryptosporidium parvum Iowa oocysts by oesophageal gavage. The intestinal microbiota of both infected (Cp+) and uninfected (Cp-) mice groups was examined by high-throughput sequencing of the bacterial 16S rDNA gene V3-V4 hypervariable region. RESULTS The most consistent change in the microbiota composition of Cp+ mice was the increased proportion of bacterial communities belonging to the Phylum Bacteroidetes. In contrast, the microbiota of Cp- mice was associated with increased proportions of several Firmicutes and Actinobacteria phyla members. CONCLUSION For the first time, our study provides evidence of an association between cryptosporidial infection and gut dysbiosis, thus contributing valuable knowledge to the as-yet little-explored field of Cryptosporidium-microbiota interactions in a neonatal mouse model.
Collapse
|
63
|
Wang S, El-Fahmawi A, Christian DA, Fang Q, Radaelli E, Chen L, Sullivan MC, Misic AM, Ellringer JA, Zhu XQ, Winter SE, Hunter CA, Beiting DP. Infection-Induced Intestinal Dysbiosis Is Mediated by Macrophage Activation and Nitrate Production. mBio 2019; 10:e00935-19. [PMID: 31138751 PMCID: PMC6538788 DOI: 10.1128/mbio.00935-19] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 04/24/2019] [Indexed: 01/08/2023] Open
Abstract
Oral infection of C57BL/6J mice with Toxoplasma gondii results in a marked bacterial dysbiosis and the development of severe pathology in the distal small intestine that is dependent on CD4+ T cells and interferon gamma (IFN-γ). This dysbiosis and bacterial translocation contribute to the development of ileal pathology, but the factors that support the bloom of bacterial pathobionts are unclear. The use of microbial community profiling and shotgun metagenomics revealed that Toxoplasma infection induces a dysbiosis dominated by Enterobacteriaceae and an increased potential for nitrate respiration. In vivo experiments using bacterial metabolic mutants revealed that during this infection, host-derived nitrate supports the expansion of Enterobacteriaceae in the ileum via nitrate respiration. Additional experiments with infected mice indicate that the IFN-γ/STAT1/iNOS axis, while essential for parasite control, also supplies a pool of nitrate that serves as a source for anaerobic respiration and supports overgrowth of Enterobacteriaceae Together, these data reveal a trade-off in intestinal immunity after oral infection of C57BL/6J mice with T. gondii, in which inducible nitric oxide synthase (iNOS) is required for parasite control, while this host enzyme is responsible for specific modification of the composition of the microbiome that contributes to pathology.IMPORTANCEToxoplasma gondii is a protozoan parasite and a leading cause of foodborne illness. Infection is initiated when the parasite invades the intestinal epithelium, and in many host species, this leads to intense inflammation and a dramatic disruption of the normal microbial ecosystem that resides in the healthy gut (the so-called microbiome). One characteristic change in the microbiome during infection with Toxoplasma-as well as numerous other pathogens-is the overgrowth of Escherichia coli or similar bacteria and a breakdown of commensal containment leading to seeding of peripheral organs with gut bacteria and subsequent sepsis. Our findings provide one clear explanation for how this process is regulated, thereby improving our understanding of the relationship between parasite infection, inflammation, and disease. Furthermore, our results could serve as the basis for the development of novel therapeutics to reduce the potential for harmful bacteria to bloom in the gut during infection.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Ayah El-Fahmawi
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David A Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Qun Fang
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Longfei Chen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Megan C Sullivan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ana M Misic
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jodi A Ellringer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Sebastian E Winter
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
64
|
Rowan-Nash AD, Korry BJ, Mylonakis E, Belenky P. Cross-Domain and Viral Interactions in the Microbiome. Microbiol Mol Biol Rev 2019; 83:e00044-18. [PMID: 30626617 PMCID: PMC6383444 DOI: 10.1128/mmbr.00044-18] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The importance of the microbiome to human health is increasingly recognized and has become a major focus of recent research. However, much of the work has focused on a few aspects, particularly the bacterial component of the microbiome, most frequently in the gastrointestinal tract. Yet humans and other animals can be colonized by a wide array of organisms spanning all domains of life, including bacteria and archaea, unicellular eukaryotes such as fungi, multicellular eukaryotes such as helminths, and viruses. As they share the same host niches, they can compete with, synergize with, and antagonize each other, with potential impacts on their host. Here, we discuss these major groups making up the human microbiome, with a focus on how they interact with each other and their multicellular host.
Collapse
Affiliation(s)
- Aislinn D Rowan-Nash
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Benjamin J Korry
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
65
|
Toro-Londono MA, Bedoya-Urrego K, Garcia-Montoya GM, Galvan-Diaz AL, Alzate JF. Intestinal parasitic infection alters bacterial gut microbiota in children. PeerJ 2019; 7:e6200. [PMID: 30643702 PMCID: PMC6327884 DOI: 10.7717/peerj.6200] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 12/03/2018] [Indexed: 12/29/2022] Open
Abstract
The study of the burden that parasites can exert upon the bacterial gut microbiota was restricted by the available technologies and their costs. Currently, next-generation sequencing coupled with traditional methodologies allows the study of eukaryotic parasites (protozoa and helminths) and its effects on the human bacterial gut microbiota diversity. This diversity can be altered by a variety of factors such as age, diet, genetics and parasitic infections among others. The disturbances of the gut microbiota have been associated with a variety of illnesses. Children population in developing countries, are especially susceptible to parasitic infections because of the lack of proper sanitation and undernutrition, allowing both, the thriving of intestinal parasites and profound alteration of the gut microbiota. In this work, we have sampled the stool of 23 children from four different children's care-centers in Medellin, Colombia, and we have identified the eukaryotic parasites by traditional and molecular methodologies coupled with microbial profiling using 16S rDNA sequencing. This mixed methodology approach has allowed us to establish an interesting relationship between Giardia intestinalis and helminth infection, having both effects upon the bacterial gut microbiota enterotypes, causing a switch from a type I to a type II enterotype upon infection.
Collapse
Affiliation(s)
- Miguel A. Toro-Londono
- Centro Nacional de Secuenciación Genómica—CNSG, Universidad de Antioquia, Medellin, Antioquia, Colombia
| | - Katherine Bedoya-Urrego
- Centro Nacional de Secuenciación Genómica—CNSG, Universidad de Antioquia, Medellin, Antioquia, Colombia
- Parasitology group, School of Medicine, Universidad de Antioquia, Medellin, Antioquia, Colombia
| | | | - Ana L. Galvan-Diaz
- Environmental Microbiology Group, School of Microbiology, Universidad de Antioquia, Medellin, Antioquia, Colombia
| | - Juan F. Alzate
- Centro Nacional de Secuenciación Genómica—CNSG, Universidad de Antioquia, Medellin, Antioquia, Colombia
- Parasitology group, School of Medicine, Universidad de Antioquia, Medellin, Antioquia, Colombia
| |
Collapse
|
66
|
Synthetic gutomics: Deciphering the microbial code for futuristic diagnosis and personalized medicine. METHODS IN MICROBIOLOGY 2019. [DOI: 10.1016/bs.mim.2019.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
67
|
Popovic A, Bourdon C, Wang PW, Guttman DS, Voskuijl W, Grigg ME, Bandsma RHJ, Parkinson J. Design and application of a novel two-amplicon approach for defining eukaryotic microbiota. MICROBIOME 2018; 6:228. [PMID: 30572961 PMCID: PMC6302507 DOI: 10.1186/s40168-018-0612-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 12/03/2018] [Indexed: 05/25/2023]
Abstract
BACKGROUND Due to a lack of systematic diagnostics, our understanding of the diversity and role of eukaryotic microbiota in human health is limited. While studies have shown fungal communities to be significant modulators of human health, information on the prevalence of taxa such as protozoa and helminths has been limited to a small number of species for which targeted molecular diagnostics are available. To probe the diversity of eukaryotic microbes and their relationships with other members of the microbiota, we applied in silico and experimental approaches to design a novel two-amplicon surveillance tool, based on sequencing regions of ribosomal RNA genes and their internal transcribed spacers. We subsequently demonstrated the utility of our approach by characterizing the eukaryotic microbiota of 46 hospitalized Malawian children suffering from Severe Acute Malnutrition (SAM). RESULTS Through in silico analysis and validation on a diverse panel of eukaryotes, we identified 18S rRNA variable genetic regions 4 and 5 (18S V4 V5), together with a region encoding 28S rRNA variable genetic region 2 and the internal transcribed spacers (transITS), as optimal for the systematic classification of eukaryotes. Sequencing of these regions revealed protozoa in all stool samples from children with SAM and helminths in most, including several eukaryotes previously implicated in malnutrition and diarrheal disease. Clinical comparisons revealed no association between protozoan parasites and diarrhea or HIV reactivity. However, the presence of Blastocystis correlated with bacterial alpha diversity and increased abundance of specific taxa, including Sporobacter, Cellulosibacter, Oscillibacter, and Roseburia. CONCLUSION We suggest this novel two-amplicon based strategy will prove an effective tool to deliver new insights into the role of eukaryotic microbiota in health and disease.
Collapse
Affiliation(s)
- Ana Popovic
- Program in Molecular Medicine, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON M5G 0A4 Canada
- Department of Biochemistry, University of Toronto, Medical Sciences Building, 1 King’s College Circle Suite 5207, Toronto, ON M5S 1A8 Canada
| | - Celine Bourdon
- Program in Translational Medicine, The Hospital for Sick Children, Peter Gilgan Center for Research and Learning, 686 Bay Street, Toronto, ON M5G 0A4 Canada
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi Coordination Centre, P.O Box 43640-00100, 197 Lenana Place 2nd Floor, Nairobi, Kenya
| | - Pauline W. Wang
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord St, Toronto, ON M5S 3G5 Canada
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, 25 Willcocks St Suite 4038, Toronto, ON M5S 3B2 Canada
| | - David S. Guttman
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord St, Toronto, ON M5S 3G5 Canada
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, 25 Willcocks St Suite 4038, Toronto, ON M5S 3B2 Canada
| | - Wieger Voskuijl
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi Coordination Centre, P.O Box 43640-00100, 197 Lenana Place 2nd Floor, Nairobi, Kenya
- Department of Pediatrics and Child Health, the College of Medicine, University of Malawi, Mahatma Gandhi, Private Bag 360, Chichiri, Blantyre, Malawi
- Global Child Health Group, Emma Children’s Hospital, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Michael E. Grigg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, 5601 Fishers Lane, MSC 9806, Bethesda, MD 20892-9806 USA
| | - Robert H. J. Bandsma
- Program in Translational Medicine, The Hospital for Sick Children, Peter Gilgan Center for Research and Learning, 686 Bay Street, Toronto, ON M5G 0A4 Canada
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi Coordination Centre, P.O Box 43640-00100, 197 Lenana Place 2nd Floor, Nairobi, Kenya
- Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, 525 University Avenue, Toronto, ON M5G 2L3 Canada
- Centre for Global Child Health, Hospital for Sick Children, 525 University Avenue Suite 702, Toronto, ON M5G 2L3 Canada
- Department of Nutritional Sciences, University of Toronto, Medical Sciences Building, 1 King’s College Circle Suite 5253A, Toronto, ON M5S 1A8 Canada
- Department of Biomedical Sciences, College of Medicine, University of Malawi, Mahatma Gandhi, Private Bag 360 Chichiri, Blantyre, Malawi
| | - John Parkinson
- Program in Molecular Medicine, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON M5G 0A4 Canada
- Department of Biochemistry, University of Toronto, Medical Sciences Building, 1 King’s College Circle Suite 5207, Toronto, ON M5S 1A8 Canada
- Department of Molecular Genetics, University of Toronto, Medical Sciences Building, 1 King’s College Circle Suite 4386, Toronto, ON M5S 1A8 Canada
| |
Collapse
|
68
|
Vonaesch P, Anderson M, Sansonetti PJ. Pathogens, microbiome and the host: emergence of the ecological Koch's postulates. FEMS Microbiol Rev 2018; 42:273-292. [PMID: 29325027 DOI: 10.1093/femsre/fuy003] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Indexed: 02/07/2023] Open
Abstract
Even though tremendous progress has been made in the last decades to elucidate the mechanisms of intestinal homeostasis, dysbiosis and disease, we are only at the beginning of understanding the complexity of the gut ecosystem and the underlying interaction networks. We are also only starting to unravel the mechanisms that pathogens have evolved to overcome the barriers imposed by the microbiota and host to exploit the system to their own benefit. Recent work in these domains clearly indicates that the 'traditional Koch's postulates', which state that a given pathogen leads to a distinct disease, are not valid for all 'infectious' diseases, but that a more complete and complex interpretation of Koch's postulates is needed in order to understand and explain them. This review summarises the current understanding of what defines a healthy gut ecosystem and highlights recent progress in uncovering the interplay between the host, its microbiota and invading intestinal pathogens. Based on these recent findings, we propose a new interpretation of Koch's postulates that we term 'ecological Koch's postulates'.
Collapse
Affiliation(s)
- Pascale Vonaesch
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 28 Rue du Dr. Roux, Paris 75015, France
| | - Mark Anderson
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 28 Rue du Dr. Roux, Paris 75015, France
| | - Philippe J Sansonetti
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 28 Rue du Dr. Roux, Paris 75015, France
| |
Collapse
|
69
|
Yu LCH. Microbiota dysbiosis and barrier dysfunction in inflammatory bowel disease and colorectal cancers: exploring a common ground hypothesis. J Biomed Sci 2018; 25:79. [PMID: 30413188 PMCID: PMC6234774 DOI: 10.1186/s12929-018-0483-8] [Citation(s) in RCA: 244] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/29/2018] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a multifactorial disease which arises as a result of the interaction of genetic, environmental, barrier and microbial factors leading to chronic inflammation in the intestine. Patients with IBD had a higher risk of developing colorectal carcinoma (CRC), of which the subset was classified as colitis-associated cancers. Genetic polymorphism of innate immune receptors had long been considered a major risk factor for IBD, and the mutations were also recently observed in CRC. Altered microbial composition (termed microbiota dybiosis) and dysfunctional gut barrier manifested by epithelial hyperpermeability and high amount of mucosa-associated bacteria were observed in IBD and CRC patients. The findings suggested that aberrant immune responses to penetrating commensal microbes may play key roles in fueling disease progression. Accumulative evidence demonstrated that mucosa-associated bacteria harbored colitogenic and protumoral properties in experimental models, supporting an active role of bacteria as pathobionts (commensal-derived opportunistic pathogens). Nevertheless, the host factors involved in bacterial dysbiosis and conversion mechanisms from lumen-dwelling commensals to mucosal pathobionts remain unclear. Based on the observation of gut leakiness in patients and the evidence of epithelial hyperpermeability prior to the onset of mucosal histopathology in colitic animals, it was postulated that the epithelial barrier dysfunction associated with mucosal enrichment of specific bacterial strains may predispose the shift to disease-associated microbiota. The speculation of leaky gut as an initiating factor for microbiota dysbiosis that eventually led to pathological consequences was proposed as the "common ground hypothesis", which will be highlighted in this review. Overall, the understanding of the core interplay between gut microbiota and epithelial barriers at early subclinical phases will shed light to novel therapeutic strategies to manage chronic inflammatory disorders and colitis-associated cancers.
Collapse
Affiliation(s)
- Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Suite 1020, #1 Jen-Ai Rd. Sec. 1, Taipei, 100, Taiwan, Republic of China.
| |
Collapse
|
70
|
Cannon MV, Bogale H, Rutt L, Humphrys M, Korpe P, Duggal P, Ravel J, Serre D. A high-throughput sequencing assay to comprehensively detect and characterize unicellular eukaryotes and helminths from biological and environmental samples. MICROBIOME 2018; 6:195. [PMID: 30373673 PMCID: PMC6206884 DOI: 10.1186/s40168-018-0581-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 10/18/2018] [Indexed: 05/10/2023]
Abstract
BACKGROUND Several of the most devastating human diseases are caused by eukaryotic parasites transmitted by arthropod vectors or through food and water contamination. These pathogens only represent a fraction of all unicellular eukaryotes and helminths that are present in the environment and many uncharacterized organisms might have subtle but pervasive effects on health, including by modifying the microbiome where they reside. Unfortunately, while we have modern molecular tools to characterize bacterial and, to a lesser extent, fungal communities, we lack suitable methods to comprehensively investigate and characterize most unicellular eukaryotes and helminths: the detection of these organisms often relies on microscopy that cannot differentiate related organisms, while molecular assays can only detect the pathogens specifically tested. RESULTS Here, we describe a novel sequencing-based assay, akin to bacterial 16S rRNA sequencing, that enables high-throughput detection and characterization of a wide range of unicellular eukaryotes and helminths, including those from taxonomical groups containing all common human parasites. We designed and evaluated taxon-specific PCR primer pairs that selectively amplify all species from eight taxonomical groups (Apicomplexa, Amoeba, Diplomonadida, Kinetoplastida, Parabasalia, Nematoda, Platyhelminthes, and Microsporidia). We then used these primers to screen DNA extracted from clinical, biological, and environmental samples, and after next-generation sequencing, identified both known and previously undescribed organisms from most taxa targeted. CONCLUSIONS This novel high-throughput assay enables comprehensive detection and identification of eukaryotic parasites and related organisms, from a wide range of complex biological and environmental samples. This approach can be easily deployed to many settings and will efficiently complement existing methods and provide a holistic perspective on the microbiome.
Collapse
Affiliation(s)
- Matthew V Cannon
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Haikel Bogale
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lindsay Rutt
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael Humphrys
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Poonum Korpe
- Department of Epidemiology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Priya Duggal
- Department of Epidemiology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David Serre
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
71
|
Abstract
Giardia is the commonest parasitic diarrheal pathogen affecting humans and a frequent cause of waterborne/foodborne parasitic diseases worldwide. Prevalence of giardiasis is higher in children, living in poor, low hygiene settings in developing countries, and in travelers returning from highly endemic areas. The clinical picture of giardiasis is heterogeneous, with high variability in severity of clinical disease. It can become chronic or be followed by post-infectious sequelae. An alarming increase in cases refractory to the conventional treatment with nitroimidazoles (ie, metronidazole) has been reported in low prevalence settings, such as European Union countries, especially in patients returning from Asia. In view of its relevance, we aim in this review to recapitulate present clinical knowledge about Giardia, with a special focus on the challenge of treatment-refractory giardiasis. We propose a working definition of clinically drug-resistant giardiasis, summarize knowledge regarding resistance mechanisms, and discuss its clinical management according to research-based evidence and medical practice. Advances in development and identification of novel drugs and potential non-pharmacological alternatives are also reviewed with the overall aim to define knowledge gaps and suggest future directions for research.
Collapse
Affiliation(s)
- Marco Lalle
- Department of Infectious Diseases, European Reference Laboratory for Parasites, Istituto Superiore di Sanità, Rome, Italy,
| | - Kurt Hanevik
- Norwegian National Advisory Unit on Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
72
|
Probiotic Product Enhances Susceptibility of Mice to Cryptosporidiosis. Appl Environ Microbiol 2018; 84:AEM.01408-18. [PMID: 30171003 DOI: 10.1128/aem.01408-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/13/2018] [Indexed: 12/11/2022] Open
Abstract
Cryptosporidiosis, a leading cause of diarrhea among infants, is caused by apicomplexan parasites classified in the genus Cryptosporidium The lack of effective drugs is motivating research to develop alternative treatments. With this aim, the impact of probiotics on the course of cryptosporidiosis was investigated. The native intestinal microbiota of specific pathogen-free immunosuppressed mice was initially depleted with orally administered antibiotics. A commercially available probiotic product intended for human consumption was subsequently added to the drinking water. Mice were infected with Cryptosporidium parvum oocysts. On average, mice treated with the probiotic product developed more severe infections. The probiotics significantly altered the fecal microbiota, but no direct association between ingestion of probiotic bacteria and their abundance in fecal microbiota was observed. These results suggest that probiotics indirectly altered the intestinal microenvironment or the intestinal epithelium in a way that favored proliferation of C. parvum IMPORTANCE The results of our study show that C. parvum responded to changes in the intestinal microenvironment induced by a nutritional supplement. This outcome paves the way for research to identify nutritional interventions aimed at limiting the impact of cryptosporidiosis.
Collapse
|
73
|
Rausch S, Midha A, Kuhring M, Affinass N, Radonic A, Kühl AA, Bleich A, Renard BY, Hartmann S. Parasitic Nematodes Exert Antimicrobial Activity and Benefit From Microbiota-Driven Support for Host Immune Regulation. Front Immunol 2018; 9:2282. [PMID: 30349532 PMCID: PMC6186814 DOI: 10.3389/fimmu.2018.02282] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/14/2018] [Indexed: 12/04/2022] Open
Abstract
Intestinal parasitic nematodes live in intimate contact with the host microbiota. Changes in the microbiome composition during nematode infection affect immune control of the parasites and shifts in the abundance of bacterial groups have been linked to the immunoregulatory potential of nematodes. Here we asked if the small intestinal parasite Heligmosomoides polygyrus produces factors with antimicrobial activity, senses its microbial environment and if the anti-nematode immune and regulatory responses are altered in mice devoid of gut microbes. We found that H. polygyrus excretory/secretory products exhibited antimicrobial activity against gram+/− bacteria. Parasites from germ-free mice displayed alterations in gene expression, comprising factors with putative antimicrobial functions such as chitinase and lysozyme. Infected germ-free mice developed increased small intestinal Th2 responses coinciding with a reduction in local Foxp3+RORγt+ regulatory T cells and decreased parasite fecundity. Our data suggest that nematodes sense their microbial surrounding and have evolved factors that limit the outgrowth of certain microbes. Moreover, the parasites benefit from microbiota-driven immune regulatory circuits, as an increased ratio of intestinal Th2 effector to regulatory T cells coincides with reduced parasite fitness in germ-free mice.
Collapse
Affiliation(s)
- Sebastian Rausch
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Ankur Midha
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Matthias Kuhring
- Bioinformatics Unit (MF 1), Robert Koch Institute, Berlin, Germany.,Core Unit Bioinformatics, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin Institute of Health Metabolomics Platform, Berlin Institute of Health (BIH), Berlin, Germany.,Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Nicole Affinass
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Aleksandar Radonic
- Centre for Biological Threats and Special Pathogens (ZBS 1), Robert Koch Institute, Berlin, Germany.,Genome Sequencing Unit (MF 2), Robert Koch Institute, Berlin, Germany
| | - Anja A Kühl
- iPATH.Berlin, Core Unit for Immunopathology for Experimental Models, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | | | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
74
|
Xu M, Jiang Z, Huang W, Yin J, Ou S, Jiang Y, Meng L, Cao S, Yu A, Cao J, Shen Y. Altered Gut Microbiota Composition in Subjects Infected With Clonorchis sinensis. Front Microbiol 2018; 9:2292. [PMID: 30323795 PMCID: PMC6172334 DOI: 10.3389/fmicb.2018.02292] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 09/07/2018] [Indexed: 12/29/2022] Open
Abstract
Clonorchiasis is an infectious disease caused by helminths of Clonorchis sinensis (C. sinensis). The adult parasite mainly inhabits the bile duct and gall bladder, and results in various complications to the hepatobiliary system. The amount of bile secreted into the intestine is reduced in cases of C. sinensis infection, which may alter the pH of the gut and decrease the amount of surfactant protein D released from the gallbladder. However, the impact of parasitic infection on the human gut microbiome remains unclear. To this end, we examined the gut microbiota composition in 47 modified Kato–Katz thick smear-positive (egg-positive) volunteers and 42 healthy controls from five rural communities. Subjects were grouped into four sub-populations based on age and infection status. High-throughput 16S rRNA gene sequencing revealed significant changes in alpha diversity between EP1 and EN1. The beta diversity showed alterations between C. sinensis-infected subjects and healthy controls. In C. sinensis infected patients, we found the significant reduction of certain taxa, such as Bacteroides and anti-inflammatory Bifidobacterium (P < 0.05). Bacteroides, a predominant gut bacteria in healthy populations, was negatively correlated with the number of C. sinensis eggs per gram (EPG, r = −0.37, P adjust < 0.01 in 20–60 years old group; r = −0.64, P adjust = 0.04 in the 60+ years old group). What’s more, the reduction in the abundance of Bifidobacterium, a common probiotic, was decreased particularly in the 60 + years old group (r = −0.50, P = 0.04). The abundance of Dorea, a potentially pro-inflammatory microbe, was higher in infected subjects than in healthy individuals (P < 0.05). Variovorax was a unique bacteria that was only detected in infected subjects. These results clearly demonstrate the significant influence of C. sinensis infection on the human gut microbiota and provided new insights into the control, prevention, diagnosis, and clinical study of clonorchiasis through the human gut microbiota.
Collapse
Affiliation(s)
- Meng Xu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China.,Chinese Center for Tropical Diseases Research, Shanghai, China.,World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, China
| | - Zhihua Jiang
- Guangxi Zhuang Autonomous Region Center for Disease Control and Prevention, Nanning, China
| | - Wen Huang
- Tengxian Center for Disease Control and Prevention, Tengxian, China
| | - Jianhai Yin
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China.,Chinese Center for Tropical Diseases Research, Shanghai, China.,World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, China
| | - Shen Ou
- Tengxian Center for Disease Control and Prevention, Tengxian, China
| | - Yanyan Jiang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China.,Chinese Center for Tropical Diseases Research, Shanghai, China.,World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, China
| | - Liyu Meng
- Tengxian Center for Disease Control and Prevention, Tengxian, China
| | - Shengkui Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China.,Chinese Center for Tropical Diseases Research, Shanghai, China.,World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, China
| | - Aiping Yu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China.,Chinese Center for Tropical Diseases Research, Shanghai, China.,World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, China
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China.,Chinese Center for Tropical Diseases Research, Shanghai, China.,World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, China
| | - Yujuan Shen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China.,Chinese Center for Tropical Diseases Research, Shanghai, China.,World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, China
| |
Collapse
|
75
|
Oriá RB, Malva JO, Foley PL, Freitas RS, Bolick DT, Guerrant RL. Revisiting Inbred Mouse Models to Study the Developing Brain: The Potential Role of Intestinal Microbiota. Front Hum Neurosci 2018; 12:358. [PMID: 30283311 PMCID: PMC6156437 DOI: 10.3389/fnhum.2018.00358] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/20/2018] [Indexed: 12/23/2022] Open
Affiliation(s)
- Reinaldo B Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and the Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza, Brazil
| | - João O Malva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Patricia L Foley
- Division of Comparative Medicine, Department of Microbiology and Immunology, Georgetown University, Washington, DC, United States
| | - Raul S Freitas
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and the Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza, Brazil
| | - David T Bolick
- Division of Infectious Diseases and International Health, Center for Global Health, University of Virginia, Charlottesville, VA, United States
| | - Richard L Guerrant
- Division of Infectious Diseases and International Health, Center for Global Health, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
76
|
Yordanova IA, Zakovic S, Rausch S, Costa G, Levashina E, Hartmann S. Micromanaging Immunity in the Murine Host vs. the Mosquito Vector: Microbiota-Dependent Immune Responses to Intestinal Parasites. Front Cell Infect Microbiol 2018; 8:308. [PMID: 30234029 PMCID: PMC6129580 DOI: 10.3389/fcimb.2018.00308] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/13/2018] [Indexed: 12/18/2022] Open
Abstract
The digestive tract plays a central role in nutrient acquisition and harbors a vast and intricate community of bacteria, fungi, viruses and parasites, collectively known as the microbiota. In recent years, there has been increasing recognition of the complex and highly contextual involvement of this microbiota in the induction and education of host innate and adaptive immune responses under homeostasis, during infection and inflammation. The gut passage and colonization by unicellular and multicellular parasite species present an immense challenge to the host immune system and to the microbial communities that provide vital support for its proper functioning. In mammals, parasitic nematodes induce distinct shifts in the intestinal microbial composition. Vice versa, the commensal microbiota has been shown to serve as a molecular adjuvant and immunomodulator during intestinal parasite infections. Moreover, similar interactions occur within insect vectors of deadly human pathogens. The gut microbiota has emerged as a crucial factor affecting vector competence in Anopheles mosquitoes, where it modulates outcomes of infections with malaria parasites. In this review, we discuss currently known involvements of the host microbiota in the instruction, support or suppression of host immune responses to gastrointestinal nematodes and protozoan parasites in mice, as well as in the malaria mosquito vector. A deeper understanding of the mechanisms underlying microbiota-dependent modulation of host and vector immunity against parasites in mammals and mosquitoes is key to a better understanding of the host-parasite relationships and the identification of more efficient approaches for intervention and treatment of parasite infections of both clinical and veterinary importance.
Collapse
Affiliation(s)
- Ivet A. Yordanova
- Center for Infection Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Suzana Zakovic
- Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Sebastian Rausch
- Center for Infection Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Giulia Costa
- Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Elena Levashina
- Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Susanne Hartmann
- Center for Infection Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
77
|
Asymptomatic Intestinal Colonization with Protist Blastocystis Is Strongly Associated with Distinct Microbiome Ecological Patterns. mSystems 2018; 3:mSystems00007-18. [PMID: 29963639 PMCID: PMC6020473 DOI: 10.1128/msystems.00007-18] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/02/2018] [Indexed: 12/25/2022] Open
Abstract
Given the results of our study and other reports of the effects of the most common human gut protist on the diversity and composition of the bacterial microbiome, Blastocystis and, possibly, other gut protists should be studied as ecosystem engineers that drive community diversity and composition. Blastocystis is the most prevalent protist of the human intestine, colonizing approximately 20% of the North American population and up to 100% in some nonindustrialized settings. Blastocystis is associated with gastrointestinal and systemic disease but can also be an asymptomatic colonizer in large populations. While recent findings in humans have shown bacterial microbiota changes associated with this protist, it is unknown whether these occur due to the presence of Blastocystis or as a result of inflammation. To explore this, we evaluated the fecal bacterial and eukaryotic microbiota in 156 asymptomatic adult subjects from a rural population in Xoxocotla, Mexico. Colonization with Blastocystis was strongly associated with an increase in bacterial alpha diversity and broad changes in beta diversity and with more discrete changes to the microbial eukaryome. More than 230 operational taxonomic units (OTUs), including those of dominant species Prevotella copri and Ruminococcus bromii, were differentially abundant in Blastocystis-colonized individuals. Large functional changes accompanied these observations, with differential abundances of 202 (out of 266) predicted metabolic pathways (PICRUSt), as well as lower fecal concentrations of acetate, butyrate, and propionate in colonized individuals. Fecal calprotectin was markedly decreased in association with Blastocystis colonization, suggesting that this ecological shift induces subclinical immune consequences to the asymptomatic host. This work is the first to show a direct association between the presence of Blastocystis and shifts in the gut bacterial and eukaryotic microbiome in the absence of gastrointestinal disease or inflammation. These results prompt further investigation of the role Blastocystis and other eukaryotes play within the human microbiome. IMPORTANCE Given the results of our study and other reports of the effects of the most common human gut protist on the diversity and composition of the bacterial microbiome, Blastocystis and, possibly, other gut protists should be studied as ecosystem engineers that drive community diversity and composition.
Collapse
|
78
|
Associations between Gut Microbiota and Common Luminal Intestinal Parasites. Trends Parasitol 2018; 34:369-377. [DOI: 10.1016/j.pt.2018.02.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/09/2018] [Accepted: 02/13/2018] [Indexed: 02/08/2023]
|
79
|
Influence of Eimeria falciformis Infection on Gut Microbiota and Metabolic Pathways in Mice. Infect Immun 2018; 86:IAI.00073-18. [PMID: 29440368 DOI: 10.1128/iai.00073-18] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 02/07/2023] Open
Abstract
Coccidiosis, caused by different species of Eimeria parasites, is an economically important disease of poultry and livestock worldwide. Here we report previously unknown alterations in the gut microbes and metabolism of BALB/c mice infected with Eimeria falciformis Specifically, we observed a significant shift in the abundance of cecal bacteria and disrupted metabolism in parasitized animals. The relative abundances of Lachnospiraceae bacterium NK4A136, Ruminiclostridium, Alistipes, and Lactobacillus declined in response to E. falciformis infection, whereas Escherichia, Shigella, Helicobacter, Klebsiella, and Bacteroides were increased. Carbohydrate and amino acid metabolites in the serum samples of infected mice were significantly altered compared to naïve controls. Levels of amino acids, including asparagine, histidine, l-cysteine, tryptophan, lysine, glycine, serine, alanine, proline, ornithine, methionine, and valine, decreased on day 7 postinfection before returning to baseline on day 14. In addition, increased levels of indolelactate and mannitol and a reduced amount of oxalic acid indicated impaired carbon metabolism upon parasitic infection. These data demonstrate that intestinal coccidial infection perturbs the microbiota and disrupts carbon and nitrogen metabolism.
Collapse
|
80
|
Pavanelli MF, Colli CM, Gomes ML, Góis MB, de Alcântara Nogueira de Melo G, de Almeida Araújo EJ, de Mello Gonçales Sant'Ana D. Comparative study of effects of assemblages AII and BIV of Giardia duodenalis on mucosa and microbiota of the small intestine in mice. Biomed Pharmacother 2018. [PMID: 29514129 DOI: 10.1016/j.biopha.2018.02.141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIMS Giardiasis is one of the major causes of diarrhea worldwide and its symptoms vary in intensity, which can be attributed to different parasite assemblages. The goal of the present study was to compare the effects of infection caused by assemblages AII and BIV ofGiardia duodenalis on the response of the small intestine, microbiota, and behavioral parameters in mice. MAIN METHODS Swiss mice were infected with assemblages AII and BIV of G. duodenalis for 15 days. Leucometry, pain, intestinal microbiota and histological parameters of the duodenum and jejunum were evaluated in the experimental groups. KEY FINDINGS Both assemblages modified the composition of the intestinal microbiota. Infection with assemblage AII promoted leukocytosis, reflected by increasing number of polymorphonuclear cells, intraepithelial lymphocytes and pain-related behavior, indicating that this was the more aggressive assemblage with regard to its effects on the intestinal mucosa and duodenum. SIGNIFICANCE The specific assemblage of the parasite is an important parameter that affects symptomatology in the host.
Collapse
|
81
|
Allain T, Chaouch S, Thomas M, Travers MA, Valle I, Langella P, Grellier P, Polack B, Florent I, Bermúdez-Humarán LG. Bile Salt Hydrolase Activities: A Novel Target to Screen Anti- Giardia Lactobacilli? Front Microbiol 2018; 9:89. [PMID: 29472903 PMCID: PMC5809405 DOI: 10.3389/fmicb.2018.00089] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/15/2018] [Indexed: 01/23/2023] Open
Abstract
Giardia duodenalis is a protozoan parasite responsible for giardiasis, a disease characterized by intestinal malabsorption, diarrhea and abdominal pain in a large number of mammal species. Giardiasis is one of the most common intestinal parasitic diseases in the world and thus a high veterinary, and public health concern. It is well-established that some probiotic bacteria may confer protection against this parasite in vitro and in vivo and we recently documented the implication of bile-salt hydrolase (BSH)-like activities from strain La1 of Lactobacillus johnsonii as mediators of these effects in vitro. We showed that these activities were able to generate deconjugated bile salts that were toxic to the parasite. In the present study, a wide collection of lactobacilli strains from different ecological origins was screened to assay their anti-giardial effects. Our results revealed that the anti-parasitic effects of some of the strains tested were well-correlated with the expression of BSH-like activities. The two most active strains in vitro, La1 and Lactobacillus gasseri CNCM I-4884, were then tested for their capacity to influence G. duodenalis infection in a suckling mice model. Strikingly, only L. gasseri CNCM I-4884 strain was able to significantly antagonize parasite growth with a dramatic reduction of the trophozoites load in the small intestine. Moreover, this strain also significantly reduced the fecal excretion of Giardia cysts after 5 days of treatment, which could contribute to blocking the transmission of the parasite, in contrast of La1 where no effect was observed. This study represents a step toward the development of new prophylactic strategies to combat G. duodenalis infection in both humans and animals.
Collapse
Affiliation(s)
- Thibault Allain
- INRA, Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, AgroParisTech, Paris, France.,UMR 7245, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Sorbonne Universités, Paris, France
| | - Soraya Chaouch
- UMR 7245, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Sorbonne Universités, Paris, France
| | - Myriam Thomas
- INRA, Ecole Nationale Vétérinaire d'Alfort, BIPAR, ENVA, ANSES, UMR, Université Paris-Est, Champs-sur-Marne, France.,INRA, Laboratoire de Santé Animale, BIPAR, ENVA, ANSES, UMR, Maisons-Alfort, France
| | - Marie-Agnès Travers
- UMR 7245, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Sorbonne Universités, Paris, France
| | - Isabelle Valle
- INRA, Ecole Nationale Vétérinaire d'Alfort, BIPAR, ENVA, ANSES, UMR, Université Paris-Est, Champs-sur-Marne, France.,INRA, Laboratoire de Santé Animale, BIPAR, ENVA, ANSES, UMR, Maisons-Alfort, France
| | - Philippe Langella
- INRA, Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, AgroParisTech, Paris, France
| | - Philippe Grellier
- UMR 7245, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Sorbonne Universités, Paris, France
| | - Bruno Polack
- INRA, Ecole Nationale Vétérinaire d'Alfort, BIPAR, ENVA, ANSES, UMR, Université Paris-Est, Champs-sur-Marne, France.,INRA, Laboratoire de Santé Animale, BIPAR, ENVA, ANSES, UMR, Maisons-Alfort, France
| | - Isabelle Florent
- UMR 7245, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Sorbonne Universités, Paris, France
| | - Luis G Bermúdez-Humarán
- INRA, Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, AgroParisTech, Paris, France
| |
Collapse
|
82
|
Cacciò SM, Lalle M, Svärd SG. Host specificity in the Giardia duodenalis species complex. INFECTION GENETICS AND EVOLUTION 2017; 66:335-345. [PMID: 29225147 DOI: 10.1016/j.meegid.2017.12.001] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/01/2017] [Accepted: 12/02/2017] [Indexed: 12/15/2022]
Abstract
Giardia duodenalis is a unicellular flagellated parasite that infects the gastrointestinal tract of a wide range of mammalian species, including humans. Investigations of protein and DNA polymorphisms revealed that G. duodenalis should be considered as a species complex, whose members, despite being morphologically indistinguishable, can be classified into eight groups, or Assemblages, separated by large genetic distances. Assemblages display various degree of host specificity, with Assemblages A and B occurring in humans and many other hosts, Assemblage C and D in canids, Assemblage E in hoofed animals, Assemblage F in cats, Assemblage G in rodents, and Assemblage H in pinnipeds. The factors determining host specificity are only partially understood, and clearly involve both the host and the parasite. Here, we review the results of in vitro and in vivo experiments, and clinical observations to highlight relevant biological and genetic differences between Assemblages, with a focus on human infection.
Collapse
Affiliation(s)
- Simone M Cacciò
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy.
| | - Marco Lalle
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Staffan G Svärd
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
83
|
Kraft MR, Klotz C, Bücker R, Schulzke JD, Aebischer T. Giardia's Epithelial Cell Interaction In Vitro: Mimicking Asymptomatic Infection? Front Cell Infect Microbiol 2017; 7:421. [PMID: 29018775 PMCID: PMC5622925 DOI: 10.3389/fcimb.2017.00421] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/12/2017] [Indexed: 12/13/2022] Open
Abstract
The protozoan parasite Giardia duodenalis is responsible for more than 280 million cases of gastrointestinal complaints ("giardiasis") every year, worldwide. Infections are acquired orally, mostly via uptake of cysts in contaminated drinking water. After transformation into the trophozoite stage, parasites start to colonize the duodenum and upper jejunum where they attach to the intestinal epithelium and replicate vegetatively. Outcome of Giardia infections vary between individuals, from self-limiting to chronic, and asymptomatic to severely symptomatic infection, with unspecific gastrointestinal complaints. One proposed mechanism for pathogenesis is the breakdown of intestinal barrier function. This has been studied by analyzing trans-epithelial electric resistances (TEER) or by indicators of epithelial permeability using labeled sugar compounds in in vitro cell culture systems, mouse models or human biopsies and epidemiological studies. Here, we discuss the results obtained mainly with epithelial cell models to highlight contradictory findings. We relate published studies to our own findings that suggest a lack of barrier compromising activities of recent G. duodenalis isolates of assemblage A, B, and E in a Caco-2 model system. We propose that this epithelial cell model be viewed as mimicking asymptomatic infection. This view will likely lead to a more informative use of the model if emphasis is shifted from aiming to identify Giardia virulence factors to defining non-parasite factors that arguably appear to be more decisive for disease.
Collapse
Affiliation(s)
- Martin R Kraft
- Unit 16 Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institute, Berlin, Germany.,Institute of Clinical Physiology, Charité Campus Benjamin Franklin, Berlin, Germany
| | - Christian Klotz
- Unit 16 Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institute, Berlin, Germany
| | - Roland Bücker
- Institute of Clinical Physiology, Charité Campus Benjamin Franklin, Berlin, Germany
| | - Jörg-Dieter Schulzke
- Institute of Clinical Physiology, Charité Campus Benjamin Franklin, Berlin, Germany
| | - Toni Aebischer
- Unit 16 Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institute, Berlin, Germany
| |
Collapse
|
84
|
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: 106] [Impact Index Per Article: 15.1] [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
|
85
|
Fink MY, Singer SM. The Intersection of Immune Responses, Microbiota, and Pathogenesis in Giardiasis. Trends Parasitol 2017; 33:901-913. [PMID: 28830665 DOI: 10.1016/j.pt.2017.08.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/18/2017] [Accepted: 08/01/2017] [Indexed: 02/07/2023]
Abstract
Giardia lamblia is one of the most common infectious protozoans in the world. Giardia rarely causes severe life-threatening diarrhea, and may even have a slight protective effect in this regard, but it is a major contributor to malnutrition and growth faltering in children in the developing world. Giardia infection also appears to be a significant risk factor for postinfectious irritable bowel and chronic fatigue syndromes. In this review we highlight recent work focused on the impact of giardiasis and the mechanisms that contribute to the various outcomes of this infection, including changes in the composition of the microbiota, activation of immune responses, and immunopathology.
Collapse
Affiliation(s)
- Marc Y Fink
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Steven M Singer
- Department of Biology, Georgetown University, Washington, DC, USA.
| |
Collapse
|
86
|
Bartelt LA, Bolick DT, Mayneris-Perxachs J, Kolling GL, Medlock GL, Zaenker EI, Donowitz J, Thomas-Beckett RV, Rogala A, Carroll IM, Singer SM, Papin J, Swann JR, Guerrant RL. Cross-modulation of pathogen-specific pathways enhances malnutrition during enteric co-infection with Giardia lamblia and enteroaggregative Escherichia coli. PLoS Pathog 2017; 13:e1006471. [PMID: 28750066 PMCID: PMC5549954 DOI: 10.1371/journal.ppat.1006471] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/14/2017] [Indexed: 12/17/2022] Open
Abstract
Diverse enteropathogen exposures associate with childhood malnutrition. To
elucidate mechanistic pathways whereby enteric microbes interact during
malnutrition, we used protein deficiency in mice to develop a new model of
co-enteropathogen enteropathy. Focusing on common enteropathogens in
malnourished children, Giardia lamblia and enteroaggregative
Escherichia coli (EAEC), we provide new insights into
intersecting pathogen-specific mechanisms that enhance malnutrition. We show for
the first time that during protein malnutrition, the intestinal microbiota
permits persistent Giardia colonization and simultaneously
contributes to growth impairment. Despite signals of intestinal injury, such as
IL1α, Giardia-infected mice lack pro-inflammatory intestinal
responses, similar to endemic pediatric Giardia infections.
Rather, Giardia perturbs microbial host co-metabolites of
proteolysis during growth impairment, whereas host nicotinamide utilization
adaptations that correspond with growth recovery increase. EAEC promotes
intestinal inflammation and markers of myeloid cell activation. During
co-infection, intestinal inflammatory signaling and cellular recruitment
responses to EAEC are preserved together with a
Giardia-mediated diminishment in myeloid cell activation.
Conversely, EAEC extinguishes markers of host energy expenditure regulatory
responses to Giardia, as host metabolic adaptations appear
exhausted. Integrating immunologic and metabolic profiles during co-pathogen
infection and malnutrition, we develop a working mechanistic model of how
cumulative diet-induced and pathogen-triggered microbial perturbations result in
an increasingly wasted host. Malnourished children are exposed to multiple sequential, and oftentimes,
persistent enteropathogens. Intestinal microbial disruption and inflammation are
known to contribute to the pathogenesis of malnutrition, but how co-pathogens
interact with each other, with the resident microbiota, or with the host to
alter these pathways is unknown. Using a new model of enteric co-infection with
Giardia lamblia and enteroaggregative Escherichia
coli in mice fed a protein deficient diet, we identify host growth
and intestinal immune responses that are differentially mediated by
pathogen-microbe interactions, including parasite-mediated changes in intestinal
microbial host co-metabolism, and altered immune responses during co-infection.
Our data model how early life cumulative enteropathogen exposures progressively
disrupt intestinal immunity and host metabolism during crucial developmental
periods. Furthermore, studies in this co-infection model reveal new insights
into environmental and microbial determinants of pathogenicity for presently
common, but poorly understood enteropathogens like Giardia
lamblia, that may not conform to existing paradigms of microbial
pathogenesis based on single pathogen-designed models.
Collapse
Affiliation(s)
- Luther A. Bartelt
- Division of Infectious Diseases, Department of Medicine, University of
North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of
America
- Center for Gastrointestinal Biology and Disease, Department of Medicine,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United
States of America
- * E-mail:
| | - David T. Bolick
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| | - Jordi Mayneris-Perxachs
- Division of Computational and Systems Medicine, Department of Surgery and
Cancer, Imperial College London, United Kingdom
| | - Glynis L. Kolling
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| | - Gregory L. Medlock
- Department of Biomedical Engineering, University of Virginia,
Charlottesville, Virginia, United States of America
| | - Edna I. Zaenker
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| | - Jeffery Donowitz
- Division of Pediatric Infectious Diseases, Children’s Hospital of
Richmond at Virginia Commonwealth University, Richmond, Virginia, United States
of America
| | - Rose Viguna Thomas-Beckett
- Division of Infectious Diseases, Department of Medicine, University of
North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of
America
| | - Allison Rogala
- Center for Gastrointestinal Biology and Disease, Department of Medicine,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United
States of America
| | - Ian M. Carroll
- Center for Gastrointestinal Biology and Disease, Department of Medicine,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United
States of America
| | - Steven M. Singer
- Department of Biology, Georgetown University, Washington, DC, United
States of America
| | - Jason Papin
- Department of Biomedical Engineering, University of Virginia,
Charlottesville, Virginia, United States of America
| | - Jonathan R. Swann
- Division of Computational and Systems Medicine, Department of Surgery and
Cancer, Imperial College London, United Kingdom
| | - Richard L. Guerrant
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| |
Collapse
|
87
|
Selber-Hnatiw S, Rukundo B, Ahmadi M, Akoubi H, Al-Bizri H, Aliu AF, Ambeaghen TU, Avetisyan L, Bahar I, Baird A, Begum F, Ben Soussan H, Blondeau-Éthier V, Bordaries R, Bramwell H, Briggs A, Bui R, Carnevale M, Chancharoen M, Chevassus T, Choi JH, Coulombe K, Couvrette F, D'Abreau S, Davies M, Desbiens MP, Di Maulo T, Di Paolo SA, Do Ponte S, Dos Santos Ribeiro P, Dubuc-Kanary LA, Duncan PK, Dupuis F, El-Nounou S, Eyangos CN, Ferguson NK, Flores-Chinchilla NR, Fotakis T, Gado Oumarou H D M, Georgiev M, Ghiassy S, Glibetic N, Grégoire Bouchard J, Hassan T, Huseen I, Ibuna Quilatan MF, Iozzo T, Islam S, Jaunky DB, Jeyasegaram A, Johnston MA, Kahler MR, Kaler K, Kamani C, Karimian Rad H, Konidis E, Konieczny F, Kurianowicz S, Lamothe P, Legros K, Leroux S, Li J, Lozano Rodriguez ME, Luponio-Yoffe S, Maalouf Y, Mantha J, McCormick M, Mondragon P, Narayana T, Neretin E, Nguyen TTT, Niu I, Nkemazem RB, O'Donovan M, Oueis M, Paquette S, Patel N, Pecsi E, Peters J, Pettorelli A, Poirier C, Pompa VR, Rajen H, Ralph RO, Rosales-Vasquez J, Rubinshtein D, Sakr S, Sebai MS, Serravalle L, Sidibe F, Sinnathurai A, Soho D, Sundarakrishnan A, Svistkova V, Ugbeye TE, Vasconcelos MS, Vincelli M, Voitovich O, Vrabel P, Wang L, Wasfi M, Zha CY, Gamberi C. Human Gut Microbiota: Toward an Ecology of Disease. Front Microbiol 2017; 8:1265. [PMID: 28769880 PMCID: PMC5511848 DOI: 10.3389/fmicb.2017.01265] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/23/2017] [Indexed: 12/17/2022] Open
Abstract
Composed of trillions of individual microbes, the human gut microbiota has adapted to the uniquely diverse environments found in the human intestine. Quickly responding to the variances in the ingested food, the microbiota interacts with the host via reciprocal biochemical signaling to coordinate the exchange of nutrients and proper immune function. Host and microbiota function as a unit which guards its balance against invasion by potential pathogens and which undergoes natural selection. Disturbance of the microbiota composition, or dysbiosis, is often associated with human disease, indicating that, while there seems to be no unique optimal composition of the gut microbiota, a balanced community is crucial for human health. Emerging knowledge of the ecology of the microbiota-host synergy will have an impact on how we implement antibiotic treatment in therapeutics and prophylaxis and how we will consider alternative strategies of global remodeling of the microbiota such as fecal transplants. Here we examine the microbiota-human host relationship from the perspective of the microbial community dynamics.
Collapse
Affiliation(s)
| | - Belise Rukundo
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Masoumeh Ahmadi
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Hayfa Akoubi
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Hend Al-Bizri
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Adelekan F Aliu
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Lilit Avetisyan
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Irmak Bahar
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Alexandra Baird
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Fatema Begum
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | - Helene Bramwell
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Alicia Briggs
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Richard Bui
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | - Talia Chevassus
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Jin H Choi
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Karyne Coulombe
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | - Meghan Davies
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Tamara Di Maulo
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | | | - Paola K Duncan
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Sara El-Nounou
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | - Tanya Fotakis
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Metodi Georgiev
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | - Tazkia Hassan
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Iman Huseen
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Tania Iozzo
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Safina Islam
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Dilan B Jaunky
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | | | - Cedric Kamani
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | - Filip Konieczny
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | - Karina Legros
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Jun Li
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | - Yara Maalouf
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Jessica Mantha
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | | | - Thi T T Nguyen
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Ian Niu
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | - Matthew Oueis
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Nehal Patel
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Emily Pecsi
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Jackie Peters
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | | | | | | | | | - Surya Sakr
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Lisa Serravalle
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Fily Sidibe
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Dominique Soho
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | | | | | - Olga Voitovich
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Pamela Vrabel
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Lu Wang
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Maryse Wasfi
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Cong Y Zha
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Chiara Gamberi
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| |
Collapse
|
88
|
Ehret T, Torelli F, Klotz C, Pedersen AB, Seeber F. Translational Rodent Models for Research on Parasitic Protozoa-A Review of Confounders and Possibilities. Front Cell Infect Microbiol 2017. [PMID: 28638807 PMCID: PMC5461347 DOI: 10.3389/fcimb.2017.00238] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Rodents, in particular Mus musculus, have a long and invaluable history as models for human diseases in biomedical research, although their translational value has been challenged in a number of cases. We provide some examples in which rodents have been suboptimal as models for human biology and discuss confounders which influence experiments and may explain some of the misleading results. Infections of rodents with protozoan parasites are no exception in requiring close consideration upon model choice. We focus on the significant differences between inbred, outbred and wild animals, and the importance of factors such as microbiota, which are gaining attention as crucial variables in infection experiments. Frequently, mouse or rat models are chosen for convenience, e.g., availability in the institution rather than on an unbiased evaluation of whether they provide the answer to a given question. Apart from a general discussion on translational success or failure, we provide examples where infections with single-celled parasites in a chosen lab rodent gave contradictory or misleading results, and when possible discuss the reason for this. We present emerging alternatives to traditional rodent models, such as humanized mice and organoid primary cell cultures. So-called recombinant inbred strains such as the Collaborative Cross collection are also a potential solution for certain challenges. In addition, we emphasize the advantages of using wild rodents for certain immunological, ecological, and/or behavioral questions. The experimental challenges (e.g., availability of species-specific reagents) that come with the use of such non-model systems are also discussed. Our intention is to foster critical judgment of both traditional and newly available translational rodent models for research on parasitic protozoa that can complement the existing mouse and rat models.
Collapse
Affiliation(s)
- Totta Ehret
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany.,Department of Molecular Parasitology, Humboldt-Universität zu BerlinBerlin, Germany
| | - Francesca Torelli
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany
| | - Christian Klotz
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany
| | - Amy B Pedersen
- School of Biological Sciences, University of EdinburghEdinburgh, United Kingdom
| | - Frank Seeber
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany
| |
Collapse
|
89
|
Pham JK, Nosala C, Scott EY, Nguyen KF, Hagen KD, Starcevich HN, Dawson SC. Transcriptomic Profiling of High-Density Giardia Foci Encysting in the Murine Proximal Intestine. Front Cell Infect Microbiol 2017; 7:227. [PMID: 28620589 PMCID: PMC5450421 DOI: 10.3389/fcimb.2017.00227] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/16/2017] [Indexed: 12/15/2022] Open
Abstract
Giardia is a highly prevalent, understudied protistan parasite causing significant diarrheal disease worldwide. Its life cycle consists of two stages: infectious cysts ingested from contaminated food or water sources, and motile trophozoites that colonize and attach to the gut epithelium, later encysting to form new cysts that are excreted into the environment. Current understanding of parasite physiology in the host is largely inferred from transcriptomic studies using Giardia grown axenically or in co-culture with mammalian cell lines. The dearth of information about the diversity of host-parasite interactions occurring within distinct regions of the gastrointestinal tract has been exacerbated by a lack of methods to directly and non-invasively interrogate disease progression and parasite physiology in live animal hosts. By visualizing Giardia infections in the mouse gastrointestinal tract using bioluminescent imaging (BLI) of tagged parasites, we recently showed that parasites colonize the gut in high-density foci. Encystation is initiated in these foci throughout the entire course of infection, yet how the physiology of parasites within high-density foci in the host gut differs from that of cells in laboratory culture is unclear. Here we use BLI to precisely select parasite samples from high-density foci in the proximal intestine to interrogate in vivo Giardia gene expression in the host. Relative to axenic culture, we noted significantly higher expression (>10-fold) of oxidative stress, membrane transporter, and metabolic and structural genes associated with encystation in the high-density foci. These differences in gene expression within parasite foci in the host may reflect physiological changes associated with high-density growth in localized regions of the gut. We also identified and verified six novel cyst-specific proteins, including new components of the cyst wall that were highly expressed in these foci. Our in vivo transcriptome data support an emerging view that parasites encyst early in localized regions in the gut, possibly as a consequence of nutrient limitation, and also impact local metabolism and physiology.
Collapse
Affiliation(s)
- Jonathan K Pham
- Department of Microbiology and Molecular Genetics, University of California, DavisDavis, CA, United States
| | - Christopher Nosala
- Department of Microbiology and Molecular Genetics, University of California, DavisDavis, CA, United States
| | - Erica Y Scott
- Department of Animal Science, University of California, DavisDavis, CA, United States
| | - Kristofer F Nguyen
- Department of Microbiology and Molecular Genetics, University of California, DavisDavis, CA, United States
| | - Kari D Hagen
- Department of Microbiology and Molecular Genetics, University of California, DavisDavis, CA, United States
| | - Hannah N Starcevich
- Department of Microbiology and Molecular Genetics, University of California, DavisDavis, CA, United States
| | - Scott C Dawson
- Department of Microbiology and Molecular Genetics, University of California, DavisDavis, CA, United States
| |
Collapse
|