Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Microb Ecol ; 87(1): 81, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829379

ABSTRACT

Koinobiont endoparasitoids regulate the physiology of their hosts through altering host immuno-metabolic responses, processes which function in tandem to shape the composition of the microbiota of these hosts. Here, we employed 16S rRNA and ITS amplicon sequencing to investigate whether parasitization by the parasitoid wasps, Diachasmimorpha longicaudata (Ashmaed) (Hymenoptera: Braconidae) and Psyttalia cosyrae (Wilkinson) (Hymenoptera: Braconidae), induces gut dysbiosis and differentially alter the gut microbial (bacteria and fungi) communities of an important horticultural pest, Bactrocera dorsalis (Hendel) (Diptera: Tephritidae). We further investigated the composition of bacterial communities of adult D. longicaudata and P. cosyrae to ascertain whether the adult parasitoids and parasitized host larvae share microbial taxa through transmission. We demonstrated that parasitism by D. longicaudata induced significant gut perturbations, resulting in the colonization and increased relative abundance of pathogenic gut bacteria. Some pathogenic bacteria like Stenotrophomonas and Morganella were detected in both the guts of D. longicaudata-parasitized B. dorsalis larvae and adult D. longicaudata wasps, suggesting a horizontal transfer of microbes from the parasitoid to the host. The bacterial community of P. cosyrae adult wasps was dominated by Arsenophonus nasoniae, whereas that of D. longicaudata adults was dominated by Paucibater spp. and Pseudomonas spp. Parasitization by either parasitoid wasp was associated with an overall reduction in fungal diversity and evenness. These findings indicate that unlike P. cosyrae which is avirulent to B. dorsalis, parasitization by D. longicaudata induces shifts in the gut bacteriome of B. dorsalis larvae to a pathobiont-dominated community. This mechanism possibly enhances its virulence against the pest, further supporting its candidacy as an effective biocontrol agent of this frugivorous tephritid fruit fly pest.


Subject(s)
Bacteria , Gastrointestinal Microbiome , Larva , RNA, Ribosomal, 16S , Tephritidae , Wasps , Animals , Tephritidae/microbiology , Tephritidae/parasitology , Wasps/microbiology , Wasps/physiology , Bacteria/genetics , Bacteria/classification , Bacteria/isolation & purification , Larva/microbiology , Larva/parasitology , Larva/growth & development , RNA, Ribosomal, 16S/genetics , Fungi/genetics , Fungi/physiology , Host-Parasite Interactions , Microbiota , Dysbiosis/microbiology , Dysbiosis/parasitology
2.
mBio ; 12(6): e0289021, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34933444

ABSTRACT

High-throughput 16S rRNA sequencing has allowed the characterization of helminth-uninfected (HU) and helminth-infected (HI) gut microbiomes, revealing distinct profiles. However, there have been no qualitative or quantitative syntheses of these studies, which show marked variation in participant age, diet, pathogen of interest, and study location. A predefined minimally biased search strategy identified 23 studies in humans. For each of these studies, we qualitatively addressed the effects of helminth infection on within-individual (alpha) and between-individual (beta) fecal microbiome diversity, infection-associated microbial taxa, the effect of helminth clearance on microbiome composition, microbiome composition as a predictor of infection status or treatment outcome, and treatment-specific effects on the fecal microbiome. Concomitantly, we performed a meta-analysis on a subset of 7 of these studies containing raw, paired-end 16S reads and individual-level metadata, comprising 424 pretreatment or untreated HI individuals and 497 HU controls. After reducing the batch effect and adjusting for age, our data demonstrated that intestinal helminth parasites can alter the host gut microbiome by increasing alpha diversity and promoting taxonomic reassortment and gradient collapse. Most strongly influencing the microbiome composition were the helminths found in the large intestine, Enterobius vermicularis and Trichuris trichiura, suggesting that this influence appears to be specific to soil-transmitted helminths (STH) species and host anatomical niche. In summary, using a large and diverse sample set captured in the meta-analysis, we were able to evaluate the influence of individual helminth species as well as species-species interactions, each of which explained a significant portion of the variation in the microbiome. IMPORTANCE The gut microbiome has established importance in regulating many aspects of human health, including nutrition and immunity. While many internal and environmental factors are known to influence the microbiome, less is known about the effects of intestinal helminth parasites (worms), which together affect one-sixth of the world's population. Through a comprehensive qualitative systematic review and quantitative meta-analysis of existing literature, we provide strong evidence that helminth infection dynamically shifts the intestinal microbiome structure. Moreover, we demonstrated that such influence seems to be specific to helminth species and host anatomical niche. Our findings suggest that the gut microbiome may underlie some of the pathology associated with intestinal worm infection and support future work to understand the precise nature of the helminth-microbiome relationship.


Subject(s)
Bacteria/classification , Bacteria/isolation & purification , Dysbiosis/microbiology , Gastrointestinal Microbiome , Helminthiasis/microbiology , Helminths/physiology , Adolescent , Adult , Aged , Animals , Bacteria/genetics , Child , Child, Preschool , Dysbiosis/parasitology , Feces/parasitology , Female , Helminthiasis/parasitology , Helminths/classification , Helminths/genetics , Humans , Infant , Male , Middle Aged , Phylogeny , Young Adult
3.
Microb Pathog ; 137: 103711, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31491548

ABSTRACT

Chagas disease affects millions of people, and it is a major cause of death in Latin America. Prevention and development of an effective treatment for this infection can be favored by a more thorough understanding of T. cruzi interaction with the microbiome of vectors and hosts. Next-generation sequencing technology vastly broadened the knowledge about intestinal bacteria composition, showing that microbiota within each host (triatomines and mammals) is composed by high diversity of species, although few dominant phyla. This fact may represent an ecological balance that was acquired during the evolutionary process of the microbiome-host complex, and that serves to perpetuate this system. In this context, commensal microbiota is also essential to protect hosts, conferring them resistance to pathogens colonization. However, in some situations, the microbiota is not able to prevent infection but only modulate it. Here we will review the role of the microbiota on the parasite-vector-host triad with a focus on the kinetoplastida of medical importance Trypanosoma cruzi. Novel strategies to control Chagas disease based on intestinal microbiome will also be discussed.


Subject(s)
Chagas Disease/microbiology , Gastrointestinal Microbiome/physiology , Insect Vectors/microbiology , Animals , Biological Evolution , Dysbiosis/microbiology , Dysbiosis/parasitology , Ecology , Host Microbial Interactions/immunology , Host-Pathogen Interactions/immunology , Humans , Trypanosoma cruzi
4.
Sci Rep ; 9(1): 11121, 2019 07 31.
Article in English | MEDLINE | ID: mdl-31366962

ABSTRACT

A plethora of data points towards a role of the gastrointestinal (GI) microbiota of neonatal and young vertebrates in supporting the development and regulation of the host immune system. However, knowledge of the impact that infections by GI helminths exert on the developing microbiota of juvenile hosts is, thus far, limited. This study investigates, for the first time, the associations between acute infections by GI helminths and the faecal microbial and metabolic profiles of a cohort of equine youngstock, prior to and following treatment with parasiticides (ivermectin). We observed that high versus low parasite burdens (measured via parasite egg counts in faecal samples) were associated with specific compositional alterations of the developing microbiome; in particular, the faecal microbiota of animals with heavy worm infection burdens was characterised by lower microbial richness, and alterations to the relative abundances of bacterial taxa with immune-modulatory functions. Amino acids and glucose were increased in faecal samples from the same cohort, which indicated the likely occurrence of intestinal malabsorption. These data support the hypothesis that GI helminth infections in young livestock are associated with significant alterations to the GI microbiota, which may impact on both metabolism and development of acquired immunity. This knowledge will direct future studies aimed to identify the long-term impact of infection-induced alterations of the GI microbiota in young livestock.


Subject(s)
Dysbiosis/microbiology , Dysbiosis/parasitology , Helminthiasis/microbiology , Helminths/parasitology , Herbivory/physiology , Horses/parasitology , Animals , Dysbiosis/drug therapy , Feces/parasitology , Female , Gastrointestinal Microbiome/physiology , Helminthiasis/drug therapy , Helminths/drug effects , Ivermectin/pharmacology , Male
5.
Front Immunol ; 10: 1423, 2019.
Article in English | MEDLINE | ID: mdl-31312200

ABSTRACT

The gastrointestinal tract hosts the largest collection of commensal microbes in the body. Infections at this site can cause significant perturbations in the microbiota, known as dysbiosis, that facilitate the expansion of pathobionts, and can elicit inappropriate immune responses that impair the intestinal barrier function. Dysbiosis typically occurs during intestinal infection with Toxoplasma gondii. Host resistance to T. gondii depends on a potent Th1 response. In addition, a Th17 response is also elicited. How Th17 cells contribute to the host response to T. gondii remains unclear. Here we show that class I-restricted T cell-associated molecule (CRTAM) expression on T cells is required for an optimal IL-17 production during T. gondii infection. Moreover, that the lack of IL-17, results in increased immunopathology caused by an impaired antimicrobial peptide production and bacterial translocation from the intestinal lumen to the mesenteric lymph nodes and spleen.


Subject(s)
Dysbiosis/parasitology , Gastrointestinal Microbiome/immunology , Immunoglobulins/metabolism , Interleukin-17/metabolism , Th17 Cells/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Cell Differentiation/genetics , Dysbiosis/immunology , Female , Immunoglobulins/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Toxoplasmosis, Animal/parasitology , alpha-Defensins/metabolism , beta-Defensins/metabolism
6.
Microbiome ; 7(1): 10, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30678738

ABSTRACT

BACKGROUND: Helminth parasites represent a significant threat to the health of human and animal populations, and there is a growing need for tools to treat, diagnose, and prevent these infections. Recent work has turned to the gut microbiome as a utilitarian agent in this regard; components of the microbiome may interact with parasites to influence their success in the gut, meaning that the microbiome may encode new anthelmintic drugs. Moreover, parasite infections may restructure the microbiome's composition in consistent ways, implying that the microbiome may be useful for diagnosing infection. The innovation of these utilities requires foundational knowledge about how parasitic infection, as well as its ultimate success in the gut and impact on the host, relates to the gut microbiome. In particular, we currently possess limited insight into how the microbiome, host pathology, and parasite burden covary during infection. Identifying interactions between these parameters may uncover novel putative methods of disrupting parasite success. RESULTS: To identify interactions between parasite success and the microbiome, we quantified longitudinal associations between an intestinal helminth of zebrafish, Pseudocapillaria tomentosa, and the gut microbiome in 210 4-month-old 5D line zebrafish. Parasite burden and parasite-associated pathology varied in severity throughout the experiment in parasite-exposed fish, with intestinal pathologic changes becoming severe at late time points. Parasite exposure, burden, and intestinal lesions were correlated with gut microbial diversity. Robust generalized linear regression identified several individual taxa whose abundance predicted parasite burden, suggesting that gut microbiota may influence P. tomentosa success. Numerous associations between taxon abundance, burden, and gut pathologic changes were also observed, indicating that the magnitude of microbiome disruption during infection varies with infection severity. Finally, a random forest classifier accurately predicted a fish's exposure to the parasite based on the abundance of gut phylotypes, which underscores the potential for using the gut microbiome to diagnose intestinal parasite infection. CONCLUSIONS: These experiments demonstrate that P. tomentosa infection disrupts zebrafish gut microbiome composition and identifies potential interactions between the gut microbiota and parasite success. The microbiome may also provide a diagnostic that would enable non-destructive passive sampling for P. tomentosa and other intestinal pathogens in zebrafish facilities.


Subject(s)
Bacteria/classification , Dysbiosis/parasitology , Gastrointestinal Microbiome/physiology , Nematoda/classification , Nematode Infections/veterinary , Zebrafish/microbiology , Zebrafish/parasitology , Animals , Female , Male , Microbial Interactions/physiology
7.
ISME J ; 11(12): 2848-2863, 2017 12.
Article in English | MEDLINE | ID: mdl-28837129

ABSTRACT

The influence of unicellular eukaryotic microorganisms on human gut health and disease is still largely unexplored. Blastocystis spp. commonly colonize the gut, but its clinical significance and ecological role are currently unsettled. We have developed a high-sensitivity bioinformatic pipeline to detect Blastocystis subtypes (STs) from shotgun metagenomics, and applied it to 12 large data sets, comprising 1689 subjects of different geographic origin, disease status and lifestyle. We confirmed and extended previous observations on the high prevalence the microrganism in the population (14.9%), its non-random and ST-specific distribution, and its ability to cause persistent (asymptomatic) colonization. These findings, along with the higher prevalence observed in non-westernized individuals, the lack of positive association with any of the disease considered, and decreased presence in individuals with dysbiosis associated with colorectal cancer and Crohn's disease, strongly suggest that Blastocystis is a component of the healthy gut microbiome. Further, we found an inverse association between body mass index and Blastocystis, and strong co-occurrence with archaeal organisms (Methanobrevibacter smithii) and several bacterial species. The association of specific microbial community structures with Blastocystis was confirmed by the high predictability (up to 0.91 area under the curve) of the microorganism colonization based on the species-level composition of the microbiome. Finally, we reconstructed and functionally profiled 43 new draft Blastocystis genomes and discovered a higher intra subtype variability of ST1 and ST2 compared with ST3 and ST4. Altogether, we provide an in-depth epidemiologic, ecological, and genomic analysis of Blastocystis, and show how metagenomics can be crucial to advance population genomics of human parasites.


Subject(s)
Blastocystis/isolation & purification , Gastrointestinal Tract/parasitology , Adolescent , Adult , Aged , Blastocystis/classification , Blastocystis/genetics , Crohn Disease/parasitology , Dysbiosis/parasitology , Feces/parasitology , Female , Humans , Male , Metagenomics , Middle Aged , Prevalence , Young Adult
8.
Cell Host Microbe ; 22(1): 1-3, 2017 Jul 12.
Article in English | MEDLINE | ID: mdl-28704646

ABSTRACT

There is increasing interest in the contribution of microbes to skin disease. In this issue of Cell Host & Microbe, Gimblet et al. (2017) demonstrate that cutaneous leishmaniasis alters the human skin microbiota. In mice, this dysbiosis is transferable to naive animals, where it augments skin inflammation and disease severity.


Subject(s)
Dysbiosis/immunology , Dysbiosis/microbiology , Dysbiosis/parasitology , Skin Diseases/immunology , Skin Diseases/microbiology , Skin Diseases/parasitology , Animals , Bacteria/classification , Bacterial Physiological Phenomena/immunology , Host-Pathogen Interactions/immunology , Humans , Inflammation/immunology , Inflammation/microbiology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/microbiology , Mice , Mice, Inbred C57BL , Microbiota/immunology , Models, Animal , Skin/immunology , Skin/microbiology , Skin/parasitology
9.
Infect Immun ; 85(6)2017 06.
Article in English | MEDLINE | ID: mdl-28396324

ABSTRACT

Giardia lamblia is the most frequently identified protozoan cause of intestinal infection. Over 200 million people are estimated to have acute or chronic giardiasis, with infection rates approaching 90% in areas where Giardia is endemic. Despite its significance in global health, the mechanisms of pathogenesis associated with giardiasis remain unclear, as the parasite neither produces a known toxin nor induces a robust inflammatory response. Giardia colonization and proliferation in the small intestine of the host may, however, disrupt the ecological homeostasis of gastrointestinal commensal microbes and contribute to diarrheal disease associated with giardiasis. To evaluate the impact of Giardia infection on the host microbiota, we used culture-independent methods to quantify shifts in the diversity of commensal microbes throughout the gastrointestinal tract in mice infected with Giardia We discovered that Giardia's colonization of the small intestine causes a systemic dysbiosis of aerobic and anaerobic commensal bacteria. Specifically, Giardia colonization is typified by both expansions in aerobic Proteobacteria and decreases in anaerobic Firmicutes and Melainabacteria in the murine foregut and hindgut. Based on these shifts, we created a quantitative index of murine Giardia-induced microbial dysbiosis. This index increased at all gut regions during the duration of infection, including both the proximal small intestine and the colon. Giardiasis could be an ecological disease, and the observed dysbiosis may be mediated directly via the parasite's unique anaerobic fermentative metabolism or indirectly via parasite induction of gut inflammation. This systemic alteration of murine gut commensal diversity may be the cause or the consequence of inflammatory and metabolic changes throughout the gut. Shifts in the commensal microbiota may explain observed variations in giardiasis between hosts with respect to host pathology, degree of parasite colonization, infection initiation, and eventual clearance.


Subject(s)
Dysbiosis/parasitology , Gastrointestinal Microbiome , Giardiasis/physiopathology , Intestine, Small/parasitology , Animals , Female , Giardia lamblia , Host-Pathogen Interactions , Intestine, Small/microbiology , Mice , Mice, Inbred C57BL , Parasite Load
10.
BMC Infect Dis ; 17(1): 58, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28073356

ABSTRACT

BACKGROUND: Bidirectional signalling between the brain and the gastrointestinal tract is regulated at neural, hormonal, and immunological levels. Recent studies have shown that helminth infections can alter the normal gut microbiota. Studies have also shown that the gut microbiota is instrumental in the normal development, maturation and function of the brain. The pathophysiological pathways by which helminth infections contribute to altered cognitive function remain poorly understood. DISCUSSION: We put forward the hypothesis that gastrointestinal infections with parasitic worms, such as helminths, induce an imbalance of the gut-brain axis, which, in turn, can detrimentally manifest in brain development. Factors supporting this hypothesis are: 1) research focusing on intelligence and school performance in school-aged children has shown helminth infections to be associated with cognitive impairment, 2) disturbances in gut microbiota have been shown to be associated with important cognitive developmental effects, and 3) helminth infections have been shown to alter the gut microbiota structure. Evidence on the complex interactions between extrinsic (parasite) and intrinsic (host-derived) factors has been synthesised and discussed. While evidence in favour of the helminth-gut microbiota-central nervous system hypothesis is circumstantial, it would be unwise to rule it out as a possible mechanism by which gastrointestinal helminth infections induce childhood cognitive morbidity. Further empirical studies are necessary to test an indirect effect of helminth infections on the modulation of mood and behaviour through its effects on the gut microbiota.


Subject(s)
Brain , Child Development , Cognitive Dysfunction/psychology , Dysbiosis/psychology , Gastrointestinal Microbiome , Helminthiasis/psychology , Intestinal Diseases, Parasitic/psychology , Adolescent , Central Nervous System , Child , Cognition , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/parasitology , Dysbiosis/microbiology , Dysbiosis/parasitology , Helminthiasis/microbiology , Humans , Intestinal Diseases, Parasitic/microbiology
11.
Arch Med Res ; 48(8): 690-700, 2017 11.
Article in English | MEDLINE | ID: mdl-29290328

ABSTRACT

The human gut is a highly complex ecosystem with an extensive microbial community, and the influence of the intestinal microbiota reaches the entire host organism. For example, the microbiome regulates fat storage, stimulates or renews epithelial cells, and influences the development and maturation of the brain and the immune system. Intestinal microbes can protect against infection by pathogenic bacteria, viruses, fungi and parasites. Hence, the maintenance of homeostasis between the gut microbiota and the rest of the body is crucial for health, with dysbiosis affecting disease. This review focuses on intestinal protozoa, especially those still representing a public health problem in Mexico, and their interactions with the microbiome and the host. The decrease in prevalence of intestinal helminthes in humans left a vacant ecological niche that was quickly occupied by protozoa. Although the mechanisms governing the interaction between intestinal microbiota and protozoa are poorly understood, it is known that the composition of the intestinal bacterial populations modulates the progression of protozoan infection and the outcome of parasitic disease. Most reports on the complex interactions between intestinal bacteria, protozoa and the immune system emphasize the protective role of the microbiota against protozoan infection. Insights into such protection may facilitate the manipulation of microbiota components to prevent and treat intestinal protozoan infections. Here we discuss recent findings about the immunoregulatory effect of intestinal microbiota with regards to intestinal colonization by protozoa, focusing on infections by Entamoeba histolytica, Blastocystis spp, Giardia duodenalis, Toxoplasma gondii and Cryptosporidium parvum. The possible consequences of the microbiota on parasitic, allergic and autoimmune disorders are also considered.


Subject(s)
Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Parasites/immunology , Parasites/microbiology , Protozoan Infections/etiology , Animals , Dysbiosis/microbiology , Dysbiosis/parasitology , Homeostasis , Humans , Mexico , Protozoan Infections/immunology , Protozoan Infections/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...