1
|
Qin X, Han J, Xi L, Zhao L, Li Z, Cui Y, Hao J. Multi-omics insights into the response of the gut microbiota and metabolites to albendazole deworming in captive Rhinopithecus brelichi. Front Microbiol 2025; 16:1581483. [PMID: 40336838 PMCID: PMC12058082 DOI: 10.3389/fmicb.2025.1581483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 04/04/2025] [Indexed: 05/09/2025] Open
Abstract
Background Parasite infection and deworming treatment affect the host gut microbiota. Exploring the response mechanism of the gut microbiota in Rhinopithecus brelichi (R. brelichi) to albendazole deworming treatment is of great value for protecting this critically endangered species. Methods and results This study used metataxonomics and metabolomics to explore the responses of the gut microbiota and metabolites of R. brelichi to albendazole deworming treatment. The results showed that deworming significantly reduced the eggs per gram of feces (EPG). The 16S rRNA gene sequencing results showed that the richness and diversity of the gut microbiota in R. brelichi after deworming were significantly increased. Meanwhile, deworming treatment also changed the composition of the gut microbiota. At the genus level, the Christensenellaceae R7 group, UCG 002, UCG 005, uncultured rumen bacterium, and Rikenellaceae RC9 gut group were significantly enriched in the pre-deworming samples. Unclassified Muribaculaceae, Prevotella 9, and Bacteroides were significantly enriched in the post-deworming samples. Metabolomics analysis revealed that the relative abundance of 382 out of 1,865 metabolites showed significant differences between the pre- and post-deworming samples. Among them, 103 metabolites were annotated based on the HMDB and mainly classified into Prenol lipids, Carboxylic acids and derivatives, and Organooxygen compounds, etc. The KEGG enrichment analysis result indicated that these metabolites were mainly involved in energy, amino acid, lipid, and purine metabolism. Correlation analysis showed that Bacteroides and unclassified Muribaculaceae, whose relative abundances were upregulated after deworming treatment, were positively correlated with Kaempferol, 5,7-Dihydroxy-3-methoxy-4'-prenyloxyflavone, Purpurin, and Rhein, which have anti-parasitic activities. The Christensenellaceae R7 group, with a downregulated relative abundance after deworming treatment, was not only negatively correlated with the above four metabolites, but also positively correlated with Retinyl beta-glucuronide, which is a storage form of vitamin A, and positively correlated with CDP-Choline, which increases the host's susceptibility to Entamoeba histolytica and Plasmodium falciparum. Conclusion This study emphasizes that deworming treatment has an impact on the gut microbiota and metabolic functions of R. brelichi. By exploiting the correlations between differential microbiota and metabolites, potential probiotics or prebiotics can be explored, thereby enhancing the efficiency of deworming and reducing its side effects.
Collapse
Affiliation(s)
- Xinxi Qin
- College of Biology and Food, Shangqiu Normal University, Shangqiu, China
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Jincheng Han
- College of Biology and Food, Shangqiu Normal University, Shangqiu, China
| | - Li Xi
- College of Biology and Food, Shangqiu Normal University, Shangqiu, China
| | - Longfei Zhao
- College of Biology and Food, Shangqiu Normal University, Shangqiu, China
| | - Zhiqiang Li
- College of Biology and Food, Shangqiu Normal University, Shangqiu, China
| | - Yanyan Cui
- College of Biology and Food, Shangqiu Normal University, Shangqiu, China
| | - Junfang Hao
- College of Biology and Food, Shangqiu Normal University, Shangqiu, China
| |
Collapse
|
2
|
Vanhove MPM, Koblmüller S, Fernandes JMO, Hahn C, Plusquin M, Kmentová N. Cichlid fishes are promising underutilized models to investigate helminth-host-microbiome interactions. Front Immunol 2025; 16:1527184. [PMID: 40018030 PMCID: PMC11864961 DOI: 10.3389/fimmu.2025.1527184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
The "Old Friends Hypothesis" suggests insufficient exposure to symbionts hinders immune development, contributing to increased immune-related diseases in the Global North. The microbiome is often the focus; helminths, potentially also offering health benefits, lack attention. Infection and effect of helminths are influenced and perhaps determined by micro-organisms. Mechanisms behind parasite-microbiome interactions are poorly understood, despite implications on host health. These interactions are typically studied for single helminth species in laboratory animal models, overlooking helminth diversity. Reviewing research on relationships between helminth and microbial diversity yielded 27 publications; most focused on human or other mammalian hosts, relying on natural exposure rather than experimental helminth inoculation. Only about half investigated host health outcomes. Remaining knowledge gaps warrant considering additional candidate model systems. Given the high helminthiasis burden and species diversity of helminths, we propose seeking models in the Global South, where a considerable proportion of research on diversity aspects of helminth-microbiome interactions took place. Low availability of genomic resources for helminths in the Global South, however, necessitates more integrative helminthological research efforts. Given substantial similarities in immune systems, several fishes are models for human health/disease. More effort could be done to establish this for cichlids, whose representatives in the African Great Lakes provide a well-delineated, closed natural system relevant to human health in view of fish-borne zoonoses and other water-borne parasites. A good baseline exists for these cichlids' genomics, parasitology, and microbiology. We suggest exploring African Great Lake cichlids as model hosts for interactions between microbial diversity, helminth diversity, and host health.
Collapse
Affiliation(s)
- Maarten P. M. Vanhove
- Research Group Zoology: Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- International Union for Conservation of Nature (IUCN) Species Survival Commission (SSC) Parasite Specialist Group, Diepenbeek, Belgium
| | | | - Jorge M. O. Fernandes
- Renewable Marine Resources Department, Institut de Ciències del Mar, Spanish National Research Council, Barcelona, Spain
| | | | - Michelle Plusquin
- Research Group Environmental Biology, Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Nikol Kmentová
- Research Group Zoology: Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- International Union for Conservation of Nature (IUCN) Species Survival Commission (SSC) Parasite Specialist Group, Diepenbeek, Belgium
- Freshwater Biology, Operational Directorate Natural Environment, Royal Belgian Institute of Natural Sciences, Brussels, Belgium
| |
Collapse
|
3
|
Appiah-Twum F, Akorli J, Okyere L, Sagoe K, Osabutey D, Cappello M, Wilson MD. The effect of single dose albendazole (400 mg) treatment on the human gut microbiome of hookworm-infected Ghanaian individuals. Sci Rep 2023; 13:11302. [PMID: 37438457 PMCID: PMC10338455 DOI: 10.1038/s41598-023-38376-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 07/07/2023] [Indexed: 07/14/2023] Open
Abstract
Microbes play a key role in human gut homeostasis, metabolic, immunologic and physiopathology of the body. A longitudinal study conducted during 2018-2021 in the Kintampo North Municipality in Ghana demonstrated low hookworm infection cure rates following treatment with a single dose of 400 mg albendazole in some communities. To investigate associations between hookworm infection and the gut microbiome, we examined stool samples from consented participants who were either cured or remained infected after treatment. At each time point, stool was collected prior to and 10-14 days after albendazole treatment. We used 16S rRNA amplicon sequencing of DNA extracted from stool samples to investigate the composition and diversity of the gut microbiota and to identify potential microbial biomarkers associated with treatment outcomes. Hookworm infection was associated with increased species richness (p = 0.0093). Among treated individuals, there was also a significant variation in microbiota composition at 10-14 days following single-dose albendazole treatment. Individuals cured of hookworm infection after treatment showed a significant reduction in microbiota composition when compared to their pre-treatment state (ANOSIM; p = 0.02), whilst individuals who failed to clear the infection showed no change in microbiota composition (ANOSIM; p = 0.35). Uninfected individuals and those who were successfully treated were similar in their microbial composition and structure. We also found that the abundance of Clostridia spp. was increased in infected individuals pre- or post-treatment. Predictive functional profiling revealed the enrichment of two pyruvate ferredoxin oxidoreductase subunit pathways in individuals who remained infected after treatment (p < 0.05), alluding to an upturn of strictly anaerobic commensal bacteria such as Clostridia spp. This study suggests a relationship between human gut microbiome dysbiosis and albendazole therapy outcomes of hookworm infection. Future studies will further characterize specific biomarkers identified within this study to establish their potential for assessment of pharmacological responses to anthelminthic therapies, as well as explore the possibility of using probiotic supplementation as an adjunct treatment to increase albendazole effectiveness against hookworm.
Collapse
Affiliation(s)
- Francis Appiah-Twum
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, PO Box LG 581, Legon, Accra, Ghana
| | - Jewelna Akorli
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, PO Box LG 581, Legon, Accra, Ghana
| | - Lydia Okyere
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, PO Box LG 581, Legon, Accra, Ghana
- Department of Pathobiology, University of Illinois, Urbana-Champaign, 2522 Vet Med Basic Sciences Bldg., 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Kate Sagoe
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, PO Box LG 581, Legon, Accra, Ghana
- Pan African University Institute for Basic Sciences, Technology, and Innovation (PAUSTI), P. O. Box 62000 00200, Nairobi, Kenya
| | - Dickson Osabutey
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, PO Box LG 581, Legon, Accra, Ghana
| | - Michael Cappello
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, 60 College St, New Haven, CT, 06520, USA
| | - Michael D Wilson
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, PO Box LG 581, Legon, Accra, Ghana.
| |
Collapse
|
4
|
Abstract
Just as mammals have coevolved with the intestinal bacterial communities that are part of the microbiota, intestinal helminths represent an important selective force on their mammalian host. The complex interaction between helminths, microbes, and their mammalian host is likely an important determinant of mutual fitness. The host immune system in particular is a critical interface with both helminths and the microbiota, and this crosstalk often determines the balance between tolerance and resistance against these widespread parasites. Hence, there are many examples of how both helminths and the microbiota can influence tissue homeostasis and homeostatic immunity. Understanding these processes at a cellular and molecular level is an exciting area of research that we seek to highlight in this review and that will potentially guide future treatment approaches.
Collapse
Affiliation(s)
- P'ng Loke
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicola L Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
Khalil MKN, Abd Razak MA, Tahir FA, Sahril N, Shahein NA, Rezali MS, Adnan MAA, Liew SH, Ab Wahab N, Shamsuddin N, Kassim MSA. Prevalence and Risk Factors of Anaemia among Orang Asli Children in Malaysia: A Scoping Review. Nutrients 2023; 15:1493. [PMID: 36986223 PMCID: PMC10053598 DOI: 10.3390/nu15061493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/08/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Anaemia continues to be a global public health burden affecting all age groups, particularly children. Indigenous people, including the Orang Asli (OA) population in Malaysia, are at risk of anaemia due to the vast disparities in social determinants of health in their population compared to the non-indigenous population. OBJECTIVES This review aimed to identify the prevalence and risk factors of anaemia among OA children in Malaysia and analyse the knowledge gaps. METHODS A systematic search was conducted in PubMed, Cochrane Library, Scopus and Google Scholar databases. This review followed Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for scoping reviews (PRISMA-ScR) guidelines. RESULTS This review identified six studies involving the participation of OA children from eight subtribes residing in Peninsular Malaysia. The overall prevalence of anaemia among OA children ranged from 21.6 to 80.0%, with iron deficiency anaemia prevalence at 34.0%. The risk factors of anaemia among OA children reported from one study in this review were being younger than ten years old children (AOR 2.11 (95% CI 1.23, 3.63)) and moderate to heavy Ascaris infections (AOR 2.05 (95% CI 1.12, 3.76)). There was no data from OA children from certain age groups and subtribes. Additionally, there is a paucity of data on risk factors for anaemia among OA children from the currently available evidence. CONCLUSION The prevalence of anaemia among OA children poses a moderate to severe public health concern. Therefore, more comprehensive studies in the future are needed to address the gaps identified in this review, primarily regarding anaemia risk factors. This data would encourage policymakers in devising effective national prevention strategies to improve morbidity and mortality among OA children in the future.
Collapse
|
6
|
Tee MZ, Er YX, Easton AV, Yap NJ, Lee IL, Devlin J, Chen Z, Ng KS, Subramanian P, Angelova A, Oyesola O, Sargsian S, Ngui R, Beiting DP, Boey CCM, Chua KH, Cadwell K, Lim YAL, Loke P, Lee SC. Gut microbiome of helminth-infected indigenous Malaysians is context dependent. MICROBIOME 2022; 10:214. [PMID: 36476263 PMCID: PMC9727879 DOI: 10.1186/s40168-022-01385-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 10/04/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND While microbiomes in industrialized societies are well characterized, indigenous populations with traditional lifestyles have microbiomes that are more akin to those of ancient humans. However, metagenomic data in these populations remains scarce, and the association with soil-transmitted helminth infection status is unclear. Here, we sequenced 650 metagenomes of indigenous Malaysians from five villages with different prevalence of helminth infections. RESULTS Individuals from villages with higher prevalences of helminth infections have more unmapped reads and greater microbial diversity. Microbial community diversity and composition were most strongly associated with different villages and the effects of helminth infection status on the microbiome varies by village. Longitudinal changes in the microbiome in response to albendazole anthelmintic treatment were observed in both helminth infected and uninfected individuals. Inference of bacterial population replication rates from origin of replication analysis identified specific replicating taxa associated with helminth infection. CONCLUSIONS Our results indicate that helminth effects on the microbiota were highly dependent on context, and effects of albendazole on the microbiota can be confounding for the interpretation of deworming studies. Furthermore, a substantial quantity of the microbiome remains unannotated, and this large dataset from an indigenous population associated with helminth infections is a valuable resource for future studies. Video Abstract.
Collapse
Affiliation(s)
- Mian Zi Tee
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Yi Xian Er
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Alice V Easton
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Nan Jiun Yap
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Ii Li Lee
- Kulliyyah of Medicine and Health Sciences, University Islam Antarabangsa Sultan Abdul Halim Mu'adzam Shah, 09300, Kuala Ketil, Kedah, Malaysia
| | - Joseph Devlin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ze Chen
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Kee Seong Ng
- Department of Gastroenterology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Poorani Subramanian
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Angelina Angelova
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Oyebola Oyesola
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Shushan Sargsian
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Romano Ngui
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Kek Heng Chua
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Ken Cadwell
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, New York University Langone Health, New York, NY, USA
| | - Yvonne Ai Lian Lim
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia.
| | - P'ng Loke
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA.
| | - Soo Ching Lee
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA.
| |
Collapse
|
7
|
Lundregan SL, Mäkinen H, Buer A, Holand H, Jensen H, Husby A. Infection by a helminth parasite is associated with changes in DNA methylation in the house sparrow. Ecol Evol 2022; 12:e9539. [PMID: 36447599 PMCID: PMC9702581 DOI: 10.1002/ece3.9539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 10/24/2022] [Accepted: 11/03/2022] [Indexed: 11/29/2022] Open
Abstract
Parasites can exert strong selective pressures on their hosts and influence the evolution of host immunity. While several studies have examined the genetic basis for parasite resistance, the role of epigenetics in the immune response to parasites is less understood. Yet, epigenetic modifications, such as changes in DNA methylation, may allow species to respond rapidly to parasite prevalence or virulence. To test the role of DNA methylation in relation to parasite infection, we examined genome-wide DNA methylation before and during infection by a parasitic nematode, Syngamus trachea, in a natural population of house sparrows (Passer domesticus) using reduced representation bisulfite sequencing (RRBS). We found that DNA methylation levels were slightly lower in infected house sparrows, and we identified candidate genes relating to the initial immune response, activation of innate and adaptive immunity, and mucus membrane functional integrity that were differentially methylated between infected and control birds. Subsequently, we used methylation-sensitive high-resolution melting (MS-HRM) analyses to verify the relationship between methylation proportion and S. trachea infection status at two candidate genes in a larger sample dataset. We found that methylation level at NR1D1, but not CLDN22, remained related to infection status and that juvenile recruitment probability was positively related to methylation level at NR1D1. This underscores the importance of performing follow-up studies on candidate genes. Our findings demonstrate that plasticity in the immune response to parasites can be epigenetically mediated and highlight the potential for epigenetic studies in natural populations to provide further mechanistic insight into host-parasite interactions.
Collapse
Affiliation(s)
- Sarah L. Lundregan
- Department of Biology, Centre for Biodiversity DynamicsNorwegian University of Science and TechnologyTrondheimNorway
| | - Hannu Mäkinen
- Department of Biology, Centre for Biodiversity DynamicsNorwegian University of Science and TechnologyTrondheimNorway
- Evolutionary Biology, Department of Ecology and GeneticsUppsala UniversityUppsalaSweden
| | - Amberly Buer
- Department of Biology, Centre for Biodiversity DynamicsNorwegian University of Science and TechnologyTrondheimNorway
| | - Håkon Holand
- Department of Biology, Centre for Biodiversity DynamicsNorwegian University of Science and TechnologyTrondheimNorway
| | - Henrik Jensen
- Department of Biology, Centre for Biodiversity DynamicsNorwegian University of Science and TechnologyTrondheimNorway
| | - Arild Husby
- Department of Biology, Centre for Biodiversity DynamicsNorwegian University of Science and TechnologyTrondheimNorway
- Evolutionary Biology, Department of Ecology and GeneticsUppsala UniversityUppsalaSweden
| |
Collapse
|
8
|
Alteration in Gut Microbiota Associated with Zinc Deficiency in School-Age Children. Nutrients 2022; 14:nu14142895. [PMID: 35889856 PMCID: PMC9319427 DOI: 10.3390/nu14142895] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 12/10/2022] Open
Abstract
Zinc deficiency could lead to a dynamic variation in gut microbial composition and function in animals. However, how zinc deficiency affects the gut microbiome in school-age children remains unclear. The purpose of this study was to profile the dynamic shifts in the gut microbiome of school-age children with zinc deficiency, and to determine whether such shifts are associated with dietary intake. A dietary survey, anthropometric measurements, and serum tests were performed on 177 school-age children, and 67 children were selected to explore the gut microbial community using amplicon sequencing. School-age children suffered from poor dietary diversity and insufficient food and nutrient intake, and 32% of them were zinc deficient. The inflammatory cytokines significantly increased in the zinc deficiency (ZD) group compared to that in the control (CK) group (p < 0.05). There was no difference in beta diversity, while the Shannon index was much higher in the ZD group (p < 0.05). At the genus level, Coprobacter, Acetivibrio, Paraprevotella, and Clostridium_XI were more abundant in the ZD group (p < 0.05). A functional predictive analysis showed that the metabolism of xenobiotics by cytochrome P450 was significantly depleted in the ZD group (p < 0.05). In conclusion, gut microbial diversity was affected by zinc deficiency with some specific bacteria highlighted in the ZD group, which may be used as biomarkers for further clinical diagnosis of zinc deficiency.
Collapse
|
9
|
Loke P, Lee SC, Oyesola OO. Effects of helminths on the human immune response and the microbiome. Mucosal Immunol 2022; 15:1224-1233. [PMID: 35732819 DOI: 10.1038/s41385-022-00532-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/17/2022] [Accepted: 05/22/2022] [Indexed: 02/04/2023]
Abstract
Helminths have evolved sophisticated immune regulating mechanisms to prevent rejection by their mammalian host. Our understanding of how the human immune system responds to these parasites remains poor compared to mouse models of infection and this limits our ability to develop vaccines as well as harness their unique properties as therapeutic strategies against inflammatory disorders. Here, we review how recent studies on human challenge infections, self-infected individuals, travelers, and endemic populations have improved our understanding of human type 2 immunity and its effects on the microbiome. The heterogeneity of responses between individuals and the limited access to tissue samples beyond the peripheral blood are challenges that limit human studies on helminths, but also provide opportunities to transform our understanding of human immunology. Organoids and single-cell sequencing are exciting new tools for immunological analysis that may aid this pursuit. Learning about the genetic and immunological basis of resistance, tolerance, and pathogenesis to helminth infections may thus uncover mechanisms that can be utilized for therapeutic purposes.
Collapse
Affiliation(s)
- P'ng Loke
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Soo Ching Lee
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Oyebola O Oyesola
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
10
|
Ianiro G, Iorio A, Porcari S, Masucci L, Sanguinetti M, Perno CF, Gasbarrini A, Putignani L, Cammarota G. How the gut parasitome affects human health. Therap Adv Gastroenterol 2022; 15:17562848221091524. [PMID: 35509426 PMCID: PMC9058362 DOI: 10.1177/17562848221091524] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/23/2022] [Indexed: 02/04/2023] Open
Abstract
The human gut microbiome (GM) is a complex ecosystem that includes numerous prokaryotic and eukaryotic inhabitants. The composition of GM can influence an array of host physiological functions including immune development. Accumulating evidence suggest that several members of non-bacterial microbiota, including protozoa and helminths, that were earlier considered as pathogens, could have a commensal or beneficial relationship with the host. Here we examine the most recent data from omics studies on prokaryota-meiofauna-host interaction as well as the impact of gut parasitome on gut bacterial ecology and its role as 'immunological driver' in health and disease to glimpse new therapeutic perspectives.
Collapse
Affiliation(s)
| | - Andrea Iorio
- Department of Diagnostic and Laboratory Medicine, Unit of Parasitology and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Serena Porcari
- Gastroenterology Unit, Fondazione Policlinico Gemelli IRCCS, Roma, Italy
| | - Luca Masucci
- Microbiology Unit, Fondazione Policlinico Universitario ‘A. Gemelli’ IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maurizio Sanguinetti
- Microbiology Unit, Fondazione Policlinico Universitario ‘A. Gemelli’ IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carlo Federico Perno
- Department of Diagnostic and Laboratory Medicine, Unit of Microbiology and Diagnostic Immunology, and Multimodal Laboratory Medicine Research Area, Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | - Antonio Gasbarrini
- Gastroenterology Unit, Fondazione Policlinico Gemelli IRCCS, Roma, Italy
| | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Unit of Parasitology and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Giovanni Cammarota
- Gastroenterology Unit, Fondazione Policlinico Gemelli IRCCS, Roma, Italy
| |
Collapse
|
11
|
Zinc Deficiency Interacts with Intestinal/Urogenital Parasites in the Pathway to Anemia in Preschool Children, Bengo–Angola. Nutrients 2022; 14:nu14071392. [PMID: 35406005 PMCID: PMC9002711 DOI: 10.3390/nu14071392] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/23/2022] [Accepted: 03/01/2022] [Indexed: 02/03/2023] Open
Abstract
In host organisms with normal micronutrient status, nutritional immunity is a strongly regulated response aiming at decreasing the progression and severity of infections. Zinc deficiency may disturb this balance, impairing immune responses to infections, which may indirectly increase infection-related anemia. Since zinc deficiency may associate directly with anemia, the role of infections is often overlooked. Herein, we investigated the participation of infections (or inflammation) in the causal pathway between zinc deficiency and anemia. This transversal study, conducted in 2015 in Bengo-Angola, enrolled 852 under-3-year-old children. Logistic regression models were used to investigate interaction and mediation effects, and significance was confirmed by the Sobel test. In sum, 6.8% of children had zinc deficiency, 45.9% had anemia, and 15.6% had at least one intestinal/urogenital parasite. Furthermore, we found (1) no evidence that inflammation mediates or interacts with zinc deficiency to cause anemia, and (2) zinc deficiency interacts with infections, significantly increasing the odds of anemia (OR: 13.26, p = 0.022). This interaction was stronger among children with iron deficiency anemia (OR: 46.66, p = 0.003). Our results suggest that zinc deficiency may impair the immune response to infections and/or that intestinal parasites could have developed mechanisms to avoid zinc-limited environments. Further studies are needed to corroborate these suggestions.
Collapse
|
12
|
Kaur R, Arora N, Nair MG, Prasad A. The interplay of helminthic neuropeptides and proteases in parasite survival and host immunomodulation. Biochem Soc Trans 2022; 50:107-118. [PMID: 35076687 PMCID: PMC9042389 DOI: 10.1042/bst20210405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 11/17/2022]
Abstract
Neuropeptides comprise a diverse and broad group of neurotransmitters in vertebrates and invertebrates, with critical roles in neuronal signal transduction. While their role in controlling learning and memory in the brains of mammals is known, their extra-synaptic function in infection and inflammation with effects on distinct tissues and immune cells is increasingly recognized. Helminth infections especially of the central nervous system (CNS), such as neurocysticercosis, induce neuropeptide production by both host and helminth, but their role in host-parasite interplay or host inflammatory response is unclear. Here, we review the neurobiology of helminths, and discuss recent studies on neuropeptide synthesis and function in the helminth as well as the host CNS and immune system. Neuropeptides are summarized according to structure and function, and we discuss the complex enzyme processing for mature neuropeptides, focusing on helminth enzymes as potential targets for novel anthelminthics. We next describe known immunomodulatory effects of mammalian neuropeptides discovered from mouse infection models and draw functional parallels with helminth neuropeptides. Last, we discuss the anti-microbial properties of neuropeptides, and how they may be involved in host-microbiota changes in helminth infection. Overall, a better understanding of the biology of helminth neuropeptides, and whether they affect infection outcomes could provide diagnostic and therapeutic opportunities for helminth infections.
Collapse
Affiliation(s)
- Rimanpreet Kaur
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh 175005, India
| | - Naina Arora
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh 175005, India
| | - Meera G. Nair
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA 92521, U.S.A
| | - Amit Prasad
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh 175005, India
| |
Collapse
|
13
|
Er YX, Lee SC, Than LTL, Muslim A, Leong KF, Kwan Z, Mohd Sayed I, Lim YAL. Tinea Imbricata among the Indigenous Communities: Current Global Epidemiology and Research Gaps Associated with Host Genetics and Skin Microbiota. J Fungi (Basel) 2022; 8:202. [PMID: 35205956 PMCID: PMC8880274 DOI: 10.3390/jof8020202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 11/17/2022] Open
Abstract
Tinea imbricata is a unique fungal skin disease that mostly affects indigenous populations in Southeast Asia, Oceania, and Central and South America. The control and management of this disease among these communities are challenging given their remote locations, certain traditional practices, and severe malnutrition status. To date, there are only a handful of reports published globally, which highlights the need for a more holistic approach in addressing this skin disease. Several bodies of evidence and reports have shown that host genetic factors have a profound influence on the pathogenesis of tinea imbricata, while skin microbiota is touted to have a role in the pathogenesis of the disease. However, there are limited studies of how host genetics and skin microbiota impact disease susceptibility in the host. To improve the understanding of this disease and to find possible long-term effective treatment among the affected indigenous communities, a comprehensive literature review is needed. Hence, this review paper aims to present the current status of tinea imbricata among the indigenous communities, together with published findings on the possible underlying reasons for its specific distribution among these communities, particularly on the ways in which host skin microbiota and host genetics affect occurrence and disease patterns. This information provides valuable insights for future research by highlighting the current knowledge gaps in these areas.
Collapse
Affiliation(s)
- Yi Xian Er
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Soo Ching Lee
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Leslie Thian-Lung Than
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Azdayanti Muslim
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, Universiti Teknologi MARA (Sungai Buloh Campus), Sungai Buloh 47000, Malaysia;
- Institute for Medical Molecular Biotechnology (IMMB), Faculty of Medicine, Universiti Teknologi MARA (Sungai Buloh Campus), Sungai Buloh 47000, Malaysia
| | - Kin Fon Leong
- Pediatric Institute, Kuala Lumpur General Hospital, Kuala Lumpur 50586, Malaysia;
| | - Zhenli Kwan
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Izandis Mohd Sayed
- Hospital Orang Asli (Aborigines) Gombak, Jalan Pahang Lama, Gombak 53100, Malaysia;
| | - Yvonne Ai-Lian Lim
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
- Centre for Malaysian Indigenous Studies, Universiti Malaya, Jalan 16/4, Seksyen 16, Petaling Jaya 46350, Malaysia
| |
Collapse
|
14
|
Helminth-Induced Human Gastrointestinal Dysbiosis: a Systematic Review and Meta-Analysis Reveals Insights into Altered Taxon Diversity and Microbial Gradient Collapse. mBio 2021; 12:e0289021. [PMID: 34933444 PMCID: PMC8689561 DOI: 10.1128/mbio.02890-21] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
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.
Collapse
|
15
|
Bancroft AJ, Grencis RK. Immunoregulatory molecules secreted by Trichuris muris. Parasitology 2021; 148:1-7. [PMID: 34075864 PMCID: PMC8660643 DOI: 10.1017/s0031182021000846] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/19/2022]
Abstract
Trichuris, whipworm nematode infections are prevalent in humans, domestic livestock and mammals. All share an epithelial dwelling niche and similar life cycle with the chronic infections that follow implying that immune evasion mechanisms are operating. Nematode excretory secretory (ES) products have been shown to be a rich source of immunomodulatory molecules for many species. The Trichuris muris model is a natural parasite of mice and has been used extensively to study host–parasite interactions and provides a tractable platform for investigation of the immunoregulatory capacity of whipworm ES. The present review details progress in identification of the composition of T. muris ES, immunomodulatory components and their potential mechanisms of action. The adult T. muris secretome is dominated by one protein with modulatory capacity although remains to be completely characterized. In addition, the secretome contains multiple other proteins and small molecules that have immunomodulatory potential, certainly by comparison to other Trichuris species. Moreover, T. muris-derived exosomes/exosome-like vesicles contain both protein and multiple miRNAs providing an alternate delivery process for molecules with the potential to modulate host immunity.
Collapse
Affiliation(s)
- Allison J. Bancroft
- Lydia Becker Institute for Immunology and Inflammation, ManchesterM13 9PT, UK
- Wellcome Trust Centre for Cell Matrix Research, Manchester M13 9PT, UK
- Division of Infection, Immunity and Respiratory Medicine, ManchesterM13 9PT, UK
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, ManchesterM13 9PL, UK
| | - Richard K. Grencis
- Lydia Becker Institute for Immunology and Inflammation, ManchesterM13 9PT, UK
- Wellcome Trust Centre for Cell Matrix Research, Manchester M13 9PT, UK
- Division of Infection, Immunity and Respiratory Medicine, ManchesterM13 9PT, UK
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, ManchesterM13 9PL, UK
| |
Collapse
|
16
|
Braseth AL, Elliott DE, Ince MN. Parasitic Infections of the Gastrointestinal Track and Liver. Gastroenterol Clin North Am 2021; 50:361-381. [PMID: 34024446 PMCID: PMC11095845 DOI: 10.1016/j.gtc.2021.02.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Parasites have coevolved with humans. Several of them colonize the human body and establish a symbiotic relationship. Other parasites cause severe and lethal diseases. Prevalence of parasitic infections is decreased in highly industrialized countries, largely due to enforced hygienic practices. In contrast, parasites cause significant morbidity and mortality in parts of the world with barriers to effective public hygiene. Some parasites have emerged as potent pathogens in specific patient populations, such as immune suppressed individuals, regardless of sanitation. This article reviews common parasites encountered in clinical practice and, in the setting of host-parasite symbiosis, discusses their immune regulatory role.
Collapse
Affiliation(s)
- Annie L Braseth
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - David E Elliott
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, Carver College of Medicine, 4546 JCP, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - M Nedim Ince
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, Carver College of Medicine, 4546 JCP, 200 Hawkins Drive, Iowa City, IA 52242, USA.
| |
Collapse
|
17
|
Rosa BA, Snowden C, Martin J, Fischer K, Kupritz J, Beshah E, Supali T, Gankpala L, Fischer PU, Urban JF, Mitreva M. Whipworm-Associated Intestinal Microbiome Members Consistent Across Both Human and Mouse Hosts. Front Cell Infect Microbiol 2021; 11:637570. [PMID: 33777847 PMCID: PMC7991909 DOI: 10.3389/fcimb.2021.637570] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
The human whipworm Trichuris trichiura infects 289 million people worldwide, resulting in substantial morbidity. Whipworm infections are difficult to treat due to low cure rates and high reinfection rates. Interactions between whipworm and its host's intestinal microbiome present a potential novel target for infection control or prevention but are very complicated and are identified using inconsistent methodology and sample types across the literature, limiting their potential usefulness. Here, we used a combined 16S rRNA gene OTU analysis approach (QIIME2) for samples from humans and mice infected with whipworm (T. trichiura and T. muris, respectively) to identify for the first time, bacterial taxa that were consistently associated with whipworm infection spanning host species and infection status using four independent comparisons (baseline infected vs uninfected and before vs after deworming for both humans and mice). Using these four comparisons, we identified significant positive associations for seven taxa including Escherichia, which has been identified to induce whipworm egg hatching, and Bacteroides, which has previously been identified as a major component of the whipworm internal microbiome. We additionally identified significant negative associations for five taxa including four members of the order Clostridiales, two from the family Lachnospiraceae, including Blautia which was previously identified as positively associated with whipworm in independent human and mouse studies. Using this approach, bacterial taxa of interest for future association and mechanistic studies were identified, and several were validated by RT-qPCR. We demonstrate the applicability of a mouse animal model for comparison to human whipworm infections with respect to whipworm-induced intestinal microbiome disruption and subsequent restoration following deworming. Overall, the novel cross-species analysis approach utilized here provides a valuable research tool for studies of the interaction between whipworm infection and the host intestinal microbiome.
Collapse
Affiliation(s)
- Bruce A. Rosa
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Caroline Snowden
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - John Martin
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Kerstin Fischer
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Jonah Kupritz
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Ethiopia Beshah
- U.S. Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD, United States
| | - Taniawati Supali
- Department of Parasitology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Lincoln Gankpala
- Public Health and Medical Research, National Public Health Institute of Liberia, Charlesville, Liberia
| | - Peter U. Fischer
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Joseph F. Urban
- U.S. Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD, United States
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
18
|
Immune Response and Microbiota Profiles during Coinfection with Plasmodium vivax and Soil-Transmitted Helminths. mBio 2020; 11:mBio.01705-20. [PMID: 33082257 PMCID: PMC7587435 DOI: 10.1128/mbio.01705-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Plasmodium (malaria) and helminth parasite coinfections are frequent, and both infections can be affected by the host gut microbiota. However, the relationship between coinfection and the gut microbiota is unclear. By performing comprehensive analyses on blood/stool samples from 130 individuals in Colombia, we found that the gut microbiota may have a stronger relationship with the number of P. vivax (malaria) parasites than with the number of helminth parasites infecting a host. Microbiota analysis identified more predictors of the P. vivax parasite burden, whereas analysis of blood samples identified predictors of the helminth parasite burden. These results were unexpected, because we expected each parasite to be associated with greater differences in its biological niche (blood for P. vivax and the intestine for helminths). Instead, we find that bacterial taxa were the strongest predictors of P. vivax parasitemia levels, while circulating TGF-β levels were the strongest predictor of helminth parasite burdens. The role of the gut microbiota during coinfection with soil-transmitted helminths (STH) and Plasmodium spp. is poorly understood. We examined peripheral blood and fecal samples from 130 individuals who were either infected with Plasmodium vivax only, coinfected with P. vivax and STH, infected with STH alone, or not infected with either P. vivax or STH. In addition to a complete blood count (CBC) with differential, transcriptional profiling of peripheral blood samples was performed by transcriptome sequencing (RNA-Seq), fecal microbial communities were determined by 16S rRNA gene sequencing, and circulating cytokine levels were measured by bead-based immunoassays. Differences in blood cell counts, including an increased percentage of neutrophils, associated with a transcriptional signature of neutrophil activation, were driven primarily by P. vivax infection. P. vivax infection was also associated with increased levels of interleukin 6 (IL-6), IL-8, and IL-10; these cytokine levels were not affected by STH coinfection. Surprisingly, P. vivax infection was more strongly associated with differences in the microbiota than STH infection. Children infected with only P. vivax exhibited elevated Bacteroides and reduced Prevotella and Clostridiaceae levels, but these differences were not observed in individuals coinfected with STH. We also observed that P. vivax parasitemia was higher in the STH-infected population. When we used machine learning to identify the most important predictors of the P. vivax parasite burden (among P. vivax-infected individuals), bacterial taxa were the strongest predictors of parasitemia. In contrast, circulating transforming growth factor β (TGF-β) was the strongest predictor of the Trichuris trichiura egg burden. This study provides unexpected evidence that the gut microbiota may have a stronger link with P. vivax than with STH infection.
Collapse
|
19
|
Kohl HM, Castillo AR, Ochoa-Repáraz J. The Microbiome as a Therapeutic Target for Multiple Sclerosis: Can Genetically Engineered Probiotics Treat the Disease? Diseases 2020; 8:diseases8030033. [PMID: 32872621 PMCID: PMC7563507 DOI: 10.3390/diseases8030033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/15/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
There is an increasing interest in the intestinal microbiota as a critical regulator of the development and function of the immune, nervous, and endocrine systems. Experimental work in animal models has provided the foundation for clinical studies to investigate associations between microbiota composition and function and human disease, including multiple sclerosis (MS). Initial work done using an animal model of brain inflammation, experimental autoimmune encephalomyelitis (EAE), suggests the existence of a microbiota-gut-brain axis connection in the context of MS, and microbiome sequence analyses reveal increases and decreases of microbial taxa in MS intestines. In this review, we discuss the impact of the intestinal microbiota on the immune system and the role of the microbiome-gut-brain axis in the neuroinflammatory disease MS. We also discuss experimental evidence supporting the hypothesis that modulating the intestinal microbiota through genetically modified probiotics may provide immunomodulatory and protective effects as a novel therapeutic approach to treat this devastating disease.
Collapse
|
20
|
Natural Infection with Giardia Is Associated with Altered Community Structure of the Human and Canine Gut Microbiome. mSphere 2020; 5:5/4/e00670-20. [PMID: 32759335 PMCID: PMC7407069 DOI: 10.1128/msphere.00670-20] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
While enteric parasitic infections are among the most important infections in lower- and middle-income countries, their impact on gut microbiota is poorly understood. We reasoned that clinical symptoms associated with these infections may be influenced by alterations of the microbiome that occur during infection. To explore this notion, we took a two-pronged approach. First, we studied a cohort of dogs naturally infected with various enteric parasites and found a strong association between parasite infection and altered gut microbiota composition. Giardia, one of the most prevalent parasite infections globally, had a particularly large impact on the microbiome. Second, we took a database-driven strategy to integrate microbiome data with clinical data from large human field studies and found that Giardia infection is also associated with marked alteration of the gut microbiome of children, suggesting a possible explanation for why Giardia has been reported to be associated with protection from moderate to severe diarrhea. Enteric parasitic infections are among the most prevalent infections in lower- and middle-income countries (LMICs) and have a profound impact on global public health. While the microbiome is increasingly recognized as a key determinant of gut health and human development, the impact of naturally acquired parasite infections on microbial community structure in the gut, and the extent to which parasite-induced changes in the microbiome may contribute to gastrointestinal symptoms, is poorly understood. Enteric parasites are routinely identified in companion animals in the United States, presenting a unique opportunity to leverage this animal model to investigate the impact of naturally acquired parasite infections on the microbiome. Clinical, parasitological, and microbiome profiling of a cohort of 258 dogs revealed a significant correlation between parasite infection and composition of the bacterial community in the gut. Relative to other enteric parasites, Giardia was associated with a more pronounced perturbation of the microbiome. To compare our findings to large-scale epidemiological studies of enteric diseases in humans, a database mining approach was employed to integrate clinical and microbiome data. Substantial and consistent alterations to microbiome structure were observed in Giardia-infected children. Importantly, infection was associated with a reduction in the relative abundance of potential pathobionts, including Gammaproteobacteria, and an increase in Prevotella—a profile often associated with gut health. Taken together, these data show that widespread Giardia infection in young animals and humans is associated with significant remodeling of the gut microbiome and provide a possible explanation for the high prevalence of asymptomatic Giardia infections observed across host species. IMPORTANCE While enteric parasitic infections are among the most important infections in lower- and middle-income countries, their impact on gut microbiota is poorly understood. We reasoned that clinical symptoms associated with these infections may be influenced by alterations of the microbiome that occur during infection. To explore this notion, we took a two-pronged approach. First, we studied a cohort of dogs naturally infected with various enteric parasites and found a strong association between parasite infection and altered gut microbiota composition. Giardia, one of the most prevalent parasite infections globally, had a particularly large impact on the microbiome. Second, we took a database-driven strategy to integrate microbiome data with clinical data from large human field studies and found that Giardia infection is also associated with marked alteration of the gut microbiome of children, suggesting a possible explanation for why Giardia has been reported to be associated with protection from moderate to severe diarrhea.
Collapse
|
21
|
Colombo SAP, Grencis RK. Immunity to Soil-Transmitted Helminths: Evidence From the Field and Laboratory Models. Front Immunol 2020; 11:1286. [PMID: 32655568 PMCID: PMC7324686 DOI: 10.3389/fimmu.2020.01286] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/21/2020] [Indexed: 12/13/2022] Open
Abstract
Infection with soil-transmitted helminths (STH) remains a major burden on global health and agriculture. Our understanding of the immunological mechanisms that govern whether an individual is resistant or susceptible to infection is derived primarily from model infections in rodents. Typically, experimental infections employ an artificially high, single bolus of parasites that leads to rapid expulsion of the primary infection and robust immunity to subsequent challenges. However, immunity in natura is generated slowly, and is only partially effective, with individuals in endemic areas retaining low-level infections throughout their lives. Therefore, there is a gap between traditional model STH systems and observations in the field. Here, we review the immune response to traditional model STH infections in the laboratory. We compare these data to studies of natural infection in humans and rodents in endemic areas, highlighting crucial differences between experimental and natural infection. We then detail the literature to date on the use of "trickle" infections to experimentally model the kinetics of natural infection.
Collapse
Affiliation(s)
- Stefano A. P. Colombo
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Richard K. Grencis
- Division of Infection, Immunity and Respiratory Medicine, Wellcome Trust Centre for Cell Matrix Research, Lydia Becker Institute for Immunology and Inflammation, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|