1
|
Wolstenholme AJ, Andersen EC, Choudhary S, Ebner F, Hartmann S, Holden-Dye L, Kashyap SS, Krücken J, Martin RJ, Midha A, Nejsum P, Neveu C, Robertson AP, von Samson-Himmelstjerna G, Walker R, Wang J, Whitehead BJ, Williams PDE. Getting around the roundworms: Identifying knowledge gaps and research priorities for the ascarids. ADVANCES IN PARASITOLOGY 2024; 123:51-123. [PMID: 38448148 PMCID: PMC11143470 DOI: 10.1016/bs.apar.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
The ascarids are a large group of parasitic nematodes that infect a wide range of animal species. In humans, they cause neglected diseases of poverty; many animal parasites also cause zoonotic infections in people. Control measures include hygiene and anthelmintic treatments, but they are not always appropriate or effective and this creates a continuing need to search for better ways to reduce the human, welfare and economic costs of these infections. To this end, Le Studium Institute of Advanced Studies organized a two-day conference to identify major gaps in our understanding of ascarid parasites with a view to setting research priorities that would allow for improved control. The participants identified several key areas for future focus, comprising of advances in genomic analysis and the use of model organisms, especially Caenorhabditis elegans, a more thorough appreciation of the complexity of host-parasite (and parasite-parasite) communications, a search for novel anthelmintic drugs and the development of effective vaccines. The participants agreed to try and maintain informal links in the future that could form the basis for collaborative projects, and to co-operate to organize future meetings and workshops to promote ascarid research.
Collapse
Affiliation(s)
- Adrian J Wolstenholme
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Université de Tours, ISP, Nouzilly, France.
| | - Erik C Andersen
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| | - Shivani Choudhary
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | - Friederike Ebner
- Department of Molecular Life Sciences, School of Life Sciences, Technische Universität München, Freising, Germany
| | - Susanne Hartmann
- Institute for Immunology, Freie Universität Berlin, Berlin, Germany
| | - Lindy Holden-Dye
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Sudhanva S Kashyap
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | - Jürgen Krücken
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Richard J Martin
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | - Ankur Midha
- Institute for Immunology, Freie Universität Berlin, Berlin, Germany
| | - Peter Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Cedric Neveu
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Université de Tours, ISP, Nouzilly, France
| | - Alan P Robertson
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | | | - Robert Walker
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Jianbin Wang
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, United States
| | | | - Paul D E Williams
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| |
Collapse
|
2
|
Fontenele ALA, de Oliveira Nóbrega CG, do Nascimento WRC, Santos PDA, de Lorena VMB, Peixoto DM, de Azevedo Albuquerque MCP, Solé D, Costa VMA, Sarinho ESC, de Souza VMO. Respiratory allergy symptoms and cytokine profiles in the presence of anti-Ascaris antibody in Giardia lamblia-infected children. Parasitol Res 2023; 122:3147-3158. [PMID: 37875615 DOI: 10.1007/s00436-023-08005-z] [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: 05/26/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
Anti-Ascaris lumbricoides (Asc) IgE and IgG can immunomodulate the allergy; however, the influence of these isotypes has not been investigated in the giardiasis and allergy. Therefore, the frequency of respiratory allergy (RA) symptoms in Giardia lamblia-infected children, with or without anti-Asc IgE, IgG1, or IgG4 and Th1, Th2/Treg, and Th17 cytokine production, was evaluated. We performed a case-control study with children aged 2-10 years old selected by questionnaire and stool exams to form the groups: infected or uninfected with RA (G-RA, n = 55; nG-RA, n = 43); infected and uninfected without RA (G-nRA, n = 59; nG-nRA, n = 54). We performed blood leukocyte counts and in vitro culture. Cytokine levels in the supernatants (CBA), serum total IgE and anti-Asc IgE (ImmunoCAP), IgG1, IgG4, and total IgA (ELISA) were measured. Infection was not associated with allergy. Infected children showed increased levels of anti-Asc IgG1, IL-2, IFN-γ, IL-4, and IL-10. There was a lower frequency of allergy-related symptoms in anti-Asc IgG1-positive children than IgG1-negative (OR = 0.38; CI = 0.17-0.90, p = 0.027) and few eosinophils in G-RA than in G-nRA and more in G-nRA than in nG-nRA, whereas TNF-α levels were higher in the G-RA than in the nG-nRA group. For infected and positive anti-Asc IgG1, there was higher TNF-α and IL-10 production than G/-IgG1. IL-10 levels were lower in nG/ + IgG1 than in infected or non-infected, and both were negative for anti-Asc IgG1. Th1/Th2/IL-10 profiles were stimulated in the infected patients, and in those with circulating anti-Asc IgG1, the TNF-α production was strengthened with a lower risk for respiratory allergy symptoms.
Collapse
Affiliation(s)
- Ana Lúcia Arruda Fontenele
- Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Recife, Brazil
| | | | | | | | | | - Décio Medeiros Peixoto
- Centro de Pesquisa em Alergia e Imunologia Clínica, Hospital das Clínicas, Universidade Federal de Pernambuco, Recife, Brazil
| | | | - Dirceu Solé
- Divisão de Alergia, Imunologia Clínica e Reumatologia, Departamento de Pediatria da, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Valdenia Maria Oliveira de Souza
- Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Recife, Brazil.
- Departamento de Ciências Farmacêuticas, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego, S/N. Cidade Universitária, 50.670-901, Recife, PE, Brazil.
| |
Collapse
|
3
|
Ahmed SA, Kotepui M, Masangkay FR, Milanez GD, Karanis P. Gastrointestinal parasites in Africa: A review. ADVANCES IN PARASITOLOGY 2023; 119:1-64. [PMID: 36707173 DOI: 10.1016/bs.apar.2022.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Data on human gastrointestinal parasites (GIP) infections in the african sub-regions and countries are mainly lacking in terms of prevalence and population stratification by afflicted age group, symptomatology, multi-parasitism, and diagnostic methods. This study aims to describe the GIP reported in african countries and discuss the extent of the burden in the african context. Only 68.42% (39/57) of african countries reported human cases of GIP with helminths (45%, CI: 40-50%, I2: 99.79%) as the predominant parasitic group infecting the african population. On a regional scale, Central Africa had the highest pooled prevalence for GIP (43%, CI: 32-54%, I2: 99.74%), while the Central African Republic led all countries with a pooled prevalence of 90% (CI: 89-92%, I2: 99.96%). The vulnerable population (patients who are minorities, children, old, poor, underfunded, or have particular medical conditions) was the most affected (50%, CI: 37-62%, I2: 99.33%), with the predominance of GIP in the 6 to <20 years age group (48%, CI: 43-54%, I2: 99.68%). Reports on multi-parasitism (44%, CI: 40-48%, I2: 99.73%) were almost double the reports of single infections (43%, CI: 27-59%, I2: 99.77%) with combined molecular and non-molecular techniques demonstrating the best performance for GIP identification. The current review spans more than 40 years of GIP reports from the african continent. Geographical characteristics, environmental factors, habits of its inhabitants, and their health status play a crucial role in GIP modulation and behaviour in its captive hosts. Strategies for regular and enhanced surveillance, policy formation, and high-level community awareness are necessary to identify the true incidence in Africa and the transmission of the pathogens via water and food.
Collapse
Affiliation(s)
- Shahira A Ahmed
- Department of Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Manas Kotepui
- Medical Technology Program, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand
| | - Frederick R Masangkay
- Department of Medical Technology, Faculty of Pharmacy, University of Santo Tomas, Manila, Philippines
| | - Giovanni D Milanez
- Department of Medical Technology, Faculty of Pharmacy, University of Santo Tomas, Manila, Philippines
| | - Panagiotis Karanis
- University of Cologne, Medical Faculty and University Hospital, Cologne, Germany; University of Nicosia Medical School, Nicosia, Cyprus.
| |
Collapse
|
4
|
Abo-Zaid MA, Hamdi AA. Evaluation of Immune Response and Haematological Parameters in Infected Male Albino Rats by Giardiasis. Parasite Immunol 2022; 44:e12908. [PMID: 35104007 DOI: 10.1111/pim.12908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 11/27/2022]
Abstract
The present work aimed to study the effects of G. lamblia infection on immunological, haematological studies and to evaluate immunoglobulins and some cytokines. Fifty male albino rats were divided into six groups. The control group including 20 rats and the infected group includes 30 rats. All the estimations were checked all over five checkpoints (CP) (7, 14, 21, 28, and 35 days post-infection). Serum levels of IgA, IgG, IgM and IgE. Cytokines INF-γ, TNF-alpha, IL-4, IL-10, and haematological parameters were determined. Cyst and trophozoite were counted. A considerable increase in the level of immunoglobulins and cytokines in all infected groups compared to the control group was documented. Furthermore, a significant decrease in red blood corpuscles, haemoglobin, and mean corpuscular haemoglobin concentration levels, whereas substantial increases in mean corpuscular volume, mean corpuscular haemoglobin and platelets were observed. Moreover, infected rats had a substantial rise in WBCs, lymphocytes, and eosinophil counts compared to the control group, whereas neutrophils and monocytes had a significant decrease. Number of trophozoites and cysts were significantly increased in infected groups before diminishing after day 28. The current results showed that Th1 and Th2 immune responses, which are characterized by the production of TNF-α, IFN-γ, IL-4 and IL-10, are important for protection against Giardia infections and also verified the balance between these cytokines and the timing of their production was crucial in G. lamblia immune response. Giardia lamblia, Immunity, Antibodies, cytokines, eosinophil.
Collapse
Affiliation(s)
- Mabrouk A Abo-Zaid
- Biology department faculty of science, Jazan University, Jazan, Saudi Arabia
| | - Aishah Ali Hamdi
- Biology department faculty of science, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
5
|
Caraballo L, Zakzuk J, Acevedo N. Helminth-derived cystatins: the immunomodulatory properties of an Ascaris lumbricoides cystatin. Parasitology 2021; 148:1-13. [PMID: 33563346 DOI: 10.1017/s0031182021000214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Helminth infections such as ascariasis elicit a type 2 immune response resembling that involved in allergic inflammation, but differing to allergy, they are also accompanied with strong immunomodulation. This has stimulated an increasing number of investigations, not only to better understand the mechanisms of allergy and helminth immunity but to find parasite-derived anti-inflammatory products that could improve the current treatments of chronic non-communicable inflammatory diseases such as asthma. A great number of helminth-derived immunomodulators have been discovered and some of them extensively analysed, showing their potential use as anti-inflammatory drugs in clinical settings. Since Ascaris lumbricoides is one of the most successful parasites, several groups have focused on the immunomodulatory properties of this helminth. As a result, several excretory/secretory components and purified molecules have been analysed, revealing interesting anti-inflammatory activities potentially useful as therapeutic tools. One of these molecules is A. lumbricoides cystatin, whose genomic, cellular, molecular, and immunomodulatory properties are described in this review.
Collapse
Affiliation(s)
- Luis Caraballo
- Institute for Immunological Research, University of Cartagena, Cartagena de Indias, Colombia
| | - Josefina Zakzuk
- Institute for Immunological Research, University of Cartagena, Cartagena de Indias, Colombia
| | - Nathalie Acevedo
- Institute for Immunological Research, University of Cartagena, Cartagena de Indias, Colombia
| |
Collapse
|
6
|
Fekete E, Allain T, Siddiq A, Sosnowski O, Buret AG. Giardia spp. and the Gut Microbiota: Dangerous Liaisons. Front Microbiol 2021; 11:618106. [PMID: 33510729 PMCID: PMC7835142 DOI: 10.3389/fmicb.2020.618106] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Alteration of the intestinal microbiome by enteropathogens is commonly associated with gastrointestinal diseases and disorders and has far-reaching consequences for overall health. Significant advances have been made in understanding the role of microbial dysbiosis during intestinal infections, including infection with the protozoan parasite Giardia duodenalis, one of the most prevalent gut protozoa. Altered species composition and diversity, functional changes in the commensal microbiota, and changes to intestinal bacterial biofilm structure have all been demonstrated during the course of Giardia infection and have been implicated in Giardia pathogenesis. Conversely, the gut microbiota has been found to regulate parasite colonization and establishment and plays a critical role in immune modulation during mono and polymicrobial infections. These disruptions to the commensal microbiome may contribute to a number of acute, chronic, and post-infectious clinical manifestations of giardiasis and may account for variations in disease presentation within and between infected populations. This review discusses recent advances in characterizing Giardia-induced bacterial dysbiosis in the gut and the roles of dysbiosis in Giardia pathogenesis.
Collapse
Affiliation(s)
- Elena Fekete
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Thibault Allain
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Affan Siddiq
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Olivia Sosnowski
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Andre G. Buret
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
7
|
Midha A, Ebner F, Schlosser-Brandenburg J, Rausch S, Hartmann S. Trilateral Relationship: Ascaris, Microbiota, and Host Cells. Trends Parasitol 2020; 37:251-262. [PMID: 33008723 DOI: 10.1016/j.pt.2020.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022]
Abstract
Ascariasis is a globally spread intestinal nematode infection of humans and a considerable concern in pig husbandry. Ascaris accomplishes a complex body migration from the intestine via the liver and lung before returning to the intestine. Tissue migration and the habitat shared with a complex microbial community pose the question of how the nematode interacts with microbes and host cells from various tissues. This review addresses the current knowledge of the trilateral relationship between Ascaris, its microbial environment, and host cells, and discusses novel approaches targeting these interactions to combat this widespread infection of livestock and man.
Collapse
Affiliation(s)
- Ankur Midha
- Institute of Immunology, Freie Universität Berlin, Robert von Ostertag-Str. 7-13, D-14163 Berlin, Germany
| | - Friederike Ebner
- Institute of Immunology, Freie Universität Berlin, Robert von Ostertag-Str. 7-13, D-14163 Berlin, Germany
| | | | - Sebastian Rausch
- Institute of Immunology, Freie Universität Berlin, Robert von Ostertag-Str. 7-13, D-14163 Berlin, Germany
| | - Susanne Hartmann
- Institute of Immunology, Freie Universität Berlin, Robert von Ostertag-Str. 7-13, D-14163 Berlin, Germany.
| |
Collapse
|
8
|
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
|
9
|
Giardia spp. promote the production of antimicrobial peptides and attenuate disease severity induced by attaching and effacing enteropathogens via the induction of the NLRP3 inflammasome. Int J Parasitol 2020; 50:263-275. [PMID: 32184085 DOI: 10.1016/j.ijpara.2019.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/12/2019] [Accepted: 12/28/2019] [Indexed: 12/14/2022]
Abstract
Polymicrobial infections of the gastro-intestinal tract are common in areas with poor sanitation. Disease outcome is the result of complex interactions between the host and pathogens. Such interactions lie at the core of future management strategies of enteric diseases. In developed countries of the world, Giardia duodenalis is a common cause of diarrheal disease. In contrast, giardiasis appears to protect children against diarrhea in countries with poor sanitation, via obscure mechanisms. We hypothesized that Giardia may protect its host from disease induced by a co-infecting pathogen such as attaching and effacing Escherichia coli. This enteropathogen is commonly implicated in pediatric diarrhea in developing countries. The findings indicate that co-infection with Giardia attenuates the severity of disease induced by Citrobacter rodentium, an equivalent of A/E E. coli in mice. Co-infection with Giardia reduced colitis, blood in stools, fecal softening, bacterial invasion, and weight loss; the protective effects were lost when co-infection occurred in Nod-like receptor pyrin-containing 3 knockout mice. In co-infected mice, elevated levels of antimicrobial peptides Murine β defensin 3 and Trefoil Factor 3, and enhanced bacterial killing, were NLRP3-dependent. Inhibition of the NLRP3 inflammasome in human enterocytes blocked the activation of AMPs and bacterial killing. The findings uncover novel NLRP3-dependent modulatory mechanisms during co-infections with Giardia spp. and A/E enteropathogens, and demonstrate how these interactions may regulate the severity of enteric disease.
Collapse
|
10
|
Oliveira YLDC, Oliveira LM, Oliveira YLM, Nascimento AMD, La Corte R, Geraldi RM, Barbosa L, Gazzinelli-Guimarães PH, Fujiwara RT, Bueno LL, Dolabella SS. Changes in the epidemiological profile of intestinal parasites after a school-based large-scale treatment for soil-transmitted helminths in a community in northeastern Brazil: Epidemiological profile after large-scale school-based treatment for STH. Acta Trop 2020; 202:105279. [PMID: 31758913 DOI: 10.1016/j.actatropica.2019.105279] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/22/2019] [Accepted: 11/19/2019] [Indexed: 11/29/2022]
Abstract
Intestinal parasites cause a significant public health problem worldwide due to the associated morbidities, mainly in infected school-aged children (SAC). The strategy of large-scale deworming in SAC to control the transmission of soil-transmitted helminths (STH) has been advocated by the World Health Organization and was recently adopted in Brazil; however, the long-term effects of mass deworming on the larger parasitological profile have been less studied. After a five-year period of school-based large-scale treatment for STH using an annual single dose of albendazole in a community of Sergipe state, Brazil, a marked reduction in prevalence was observed (15.4%% vs.7.4% for Ascaris sp., 6.0%% vs. 0.4% for hookworm, and 12.8%% vs. 4.5%% for Trichuris trichiura), with the exception of Strongyloides stercoralis, which had no statistically significant change in prevalence. There was, however, an increase in the prevalence of intestinal protozoans, specifically Entamoeba histolytica/E. dispar (0.0%% vs. 36.0%), Blastocystis hominis (0.0%% vs. 40.1%), and Giardia duodenalis (5.6%% vs. 14.5%). Although the findings showed a dramatic reduction in the prevalence of STH after four rounds of preventive chemotherapy, there was an increase in intestinal protozoan infections, indicating a change in the epidemiological profile.
Collapse
Affiliation(s)
- Yvanna L D C Oliveira
- Department of Morphology, Center of Biology and Health Sciences, Universidade Federal de Sergipe, São Cristovão, Sergipe 49100-000, Brazil.
| | - Luciana M Oliveira
- Department of Morphology, Center of Biology and Health Sciences, Universidade Federal de Sergipe, São Cristovão, Sergipe 49100-000, Brazil.
| | - Yrna L M Oliveira
- Department of Morphology, Center of Biology and Health Sciences, Universidade Federal de Sergipe, São Cristovão, Sergipe 49100-000, Brazil.
| | - Ana M D Nascimento
- Department of Morphology, Center of Biology and Health Sciences, Universidade Federal de Sergipe, São Cristovão, Sergipe 49100-000, Brazil.
| | - Roseli La Corte
- Department of Morphology, Center of Biology and Health Sciences, Universidade Federal de Sergipe, São Cristovão, Sergipe 49100-000, Brazil.
| | - Ricardo M Geraldi
- Department of Morphology, Center of Biology and Health Sciences, Universidade Federal de Sergipe, São Cristovão, Sergipe 49100-000, Brazil.
| | - Luciene Barbosa
- Department of Morphology, Center of Biology and Health Sciences, Universidade Federal de Sergipe, São Cristovão, Sergipe 49100-000, Brazil.
| | - Pedro H Gazzinelli-Guimarães
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Ricardo T Fujiwara
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Lilian L Bueno
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Silvio S Dolabella
- Department of Morphology, Center of Biology and Health Sciences, Universidade Federal de Sergipe, São Cristovão, Sergipe 49100-000, Brazil.
| |
Collapse
|
11
|
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
|
12
|
Uiterwijk M, Nijsse R, Kooyman FNJ, Wagenaar JA, Mughini-Gras L, Ploeger HW. Host factors associated with Giardia duodenalis infection in dogs across multiple diagnostic tests. Parasit Vectors 2019; 12:556. [PMID: 31752993 PMCID: PMC6873540 DOI: 10.1186/s13071-019-3810-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/15/2019] [Indexed: 12/03/2022] Open
Abstract
Background The aim of this study was to assess potential associations between Giardia duodenalis infection in dogs, as determined by three diagnostic tests, and dog’s group of origin, fecal consistency, age, sex, neuter status, and co-infections with other gastrointestinal parasites. Methods Fecal samples from 1291 dogs from four groups (household, shelter, hunting and clinical dogs) were tested with qPCR, rapid enzyme immunochromatographic assay (IDEXX SNAP®Giardia), and direct immunofluorescence (DFA, Merifluor) for presence of G. duodenalis. Moreover, fecal samples were tested with centrifugation sedimentation flotation (CSF) coproscopical analysis for presence of gastrointestinal parasites. Associations were expressed as odds ratios (ORs). Results Several significant associations were found, of which a few were consistent for all three tests and Giardia positivity in general (positive with at least one of these tests). Dogs older than one year were significantly less likely to test positive for Giardia than younger dogs. Group-housed dogs, especially hunting dogs, were significantly more likely to test positive for Giardia compared to household and clinical dogs. A consistently significant association with Trichuris appeared to be driven by the high prevalence in hunting dogs. Although there was no significant association between loose stool and Giardia infection in the overall population, household dogs were significantly more likely to test Giardia-positive when having loose stool. Overall, Giardia-positive dogs with loose stool shed significantly more cysts, both determined semi-quantitatively with CSF and quantitatively by qPCR, than positive dogs with no loose stool. When other gastrointestinal parasites were present, significantly fewer cysts were detected with CSF, but this was not confirmed with qPCR. Conclusion Giardia is the most common gastrointestinal parasite in Dutch dogs, except for hunting dogs, in which Trichuris and strongyle-type eggs (hookworms) prevailed. Giardia infection was not significantly associated with loose stool, except for household dogs. Young dogs and group-housed dogs were significantly more often Giardia-positive. These associations were consistent across diagnostic tests. Young dogs, clinical dogs and dogs with loose stool shed Giardia cysts in the highest numbers. If another gastrointestinal parasite was present lower numbers of cysts were observed by microscope (CSF), but not with a molecular method (qPCR).
Collapse
Affiliation(s)
- Mathilde Uiterwijk
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,The Netherlands Food and Consumer Product Safety Authority (NVWA), Centre Monitoring Vectors, Wageningen, The Netherlands
| | - Rolf Nijsse
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Frans N J Kooyman
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jaap A Wagenaar
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Lapo Mughini-Gras
- National Institute for Public Health and the Environment (RIVM), Centre for Infectious Disease Control (CIb), Bilthoven, The Netherlands.,Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Harm W Ploeger
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
13
|
Aw JYH, Clarke NE, McCarthy JS, Traub RJ, Amaral S, Huque MH, Andrews RM, Gray DJ, Clements ACA, Vaz Nery S. Giardia duodenalis infection in the context of a community-based deworming and water, sanitation and hygiene trial in Timor-Leste. Parasit Vectors 2019; 12:491. [PMID: 31627736 PMCID: PMC6798381 DOI: 10.1186/s13071-019-3752-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/14/2019] [Indexed: 12/26/2022] Open
Abstract
Background Giardiasis is a common diarrhoeal disease caused by the protozoan Giardia duodenalis. It is prevalent in low-income countries in the context of inadequate access to water, sanitation and hygiene (WASH), and is frequently co-endemic with neglected tropical diseases such as soil-transmitted helminth (STH) infections. Large-scale periodic deworming programmes are often implemented in these settings; however, there is limited evidence for the impact of regular anthelminthic treatment on G. duodenalis infection. Additionally, few studies have examined the impact of WASH interventions on G. duodenalis. Methods The WASH for WORMS cluster randomised controlled trial was conducted in remote communities in Manufahi municipality, Timor-Leste, between 2012 and 2016. All study communities received four rounds of deworming with albendazole at six-monthly intervals. Half were randomised to additionally receive a community-level WASH intervention following study baseline. We measured G. duodenalis infection in study participants every six months for two years, immediately prior to deworming, as a pre-specified secondary outcome of the trial. WASH access and behaviours were measured using questionnaires. Results There was no significant change in G. duodenalis prevalence in either study arm between baseline and the final study follow-up. We found no additional benefit of the community-level WASH intervention on G. duodenalis infection (relative risk: 1.05, 95% CI: 0.72–1.54). Risk factors for G. duodenalis infection included living in a household with a child under five years of age (adjusted odds ratio, aOR: 1.35, 95% CI: 1.04–1.75), living in a household with more than six people (aOR: 1.32, 95% CI: 1.02–1.72), and sampling during the rainy season (aOR: 1.23, 95% CI: 1.04–1.45). Individuals infected with the hookworm Necator americanus were less likely to have G. duodenalis infection (aOR: 0.71, 95% CI: 0.57–0.88). Conclusions Prevalence of G. duodenalis was not affected by a community WASH intervention or by two years of regular deworming with albendazole. Direct household contacts appear to play a dominant role in driving transmission. We found evidence of antagonistic effects between G. duodenalis and hookworm infection, which warrants further investigation in the context of global deworming efforts. Trial registration Australian New Zealand Clinical Trials Registry, ACTRN12614000680662. Registered 27 June 2014, retrospectively registered. https://anzctr.org.au/Trial/Registration/TrialReview.aspx?id=366540.![]()
Collapse
Affiliation(s)
- Jessica Y H Aw
- Research School of Population Health, Australian National University, Canberra, ACT, Australia
| | - Naomi E Clarke
- Research School of Population Health, Australian National University, Canberra, ACT, Australia. .,The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, NSW, Australia.
| | - James S McCarthy
- Clinical Tropical Medicine Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Rebecca J Traub
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Salvador Amaral
- Research School of Population Health, Australian National University, Canberra, ACT, Australia
| | - Md Hamidul Huque
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, NSW, Australia
| | - Ross M Andrews
- Research School of Population Health, Australian National University, Canberra, ACT, Australia.,Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Darren J Gray
- Research School of Population Health, Australian National University, Canberra, ACT, Australia
| | - Archie C A Clements
- Research School of Population Health, Australian National University, Canberra, ACT, Australia.,Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Susana Vaz Nery
- Research School of Population Health, Australian National University, Canberra, ACT, Australia.,The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
14
|
Coronado S, Zakzuk J, Regino R, Ahumada V, Benedetti I, Angelina A, Palomares O, Caraballo L. Ascaris lumbricoides Cystatin Prevents Development of Allergic Airway Inflammation in a Mouse Model. Front Immunol 2019; 10:2280. [PMID: 31611876 PMCID: PMC6777510 DOI: 10.3389/fimmu.2019.02280] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/09/2019] [Indexed: 12/17/2022] Open
Abstract
Severe helminth infections are negatively associated to allergic diseases like asthma; therefore, the immunomodulatory properties of parasite-derived components have been analyzed, raising the possibility of their use as anti-inflammatory molecules. We evaluated the immunomodulatory properties of Ascaris lumbricoides recombinant cysteine protease inhibitor (rAl-CPI) in a mouse model of allergic airway inflammation induced by the house dust mite (HDM) Blomia tropicalis and its effects on human monocyte-derived dendritic cells (HmoDCs). The B. tropicalis sensitized/challenged mice developed extensive cellular airway inflammatory response, which was significantly reduced upon treatment with rAl-CPI prior to B. tropicalis sensitization, affecting particularly the perivascular/peribronchial infiltrate cells, eosinophils/neutrophils, and goblet cells. A significant decrease of Th2 cytokines, total, and specific IgE antibodies was observed in rAl-CPI treated mice. The antibody response was biased to IgG, mainly IgG2a. Administration of rAl-CPI-alone and rAl-CPI before mite sensitization were associated with a significant increase of regulatory T cells (Tregs) in spleen and elevated IL-10 levels in BAL and splenocytes culture supernatants, which was partially affected by anti-IL10 receptor use. In vitro, rAl-CPI showed a modulatory effect on HmoDCs, lowering the expression of HLA-DR, CD83, and CD86, while inducing IL-10 and IL-6 production. This suggests an inhibition of HmoDC maturation and a possible link with the inhibition of the allergic response observed in the murine model.
Collapse
Affiliation(s)
- Sandra Coronado
- Institute for Immunological Research, Universidad de Cartagena, Cartagena, Colombia
| | - Josefina Zakzuk
- Institute for Immunological Research, Universidad de Cartagena, Cartagena, Colombia
| | - Ronald Regino
- Institute for Immunological Research, Universidad de Cartagena, Cartagena, Colombia
| | - Velky Ahumada
- Institute for Immunological Research, Universidad de Cartagena, Cartagena, Colombia
| | - Ines Benedetti
- Faculty of Medicine, Universidad de Cartagena, Cartagena, Colombia
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology, Chemistry School, Complutense University of Madrid, Madrid, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, Chemistry School, Complutense University of Madrid, Madrid, Spain
| | - Luis Caraballo
- Institute for Immunological Research, Universidad de Cartagena, Cartagena, Colombia
| |
Collapse
|
15
|
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
|
16
|
Chard AN, Baker KK, Tsai K, Levy K, Sistrunk JR, Chang HH, Freeman MC. Associations between soil-transmitted helminthiasis and viral, bacterial, and protozoal enteroinfections: a cross-sectional study in rural Laos. Parasit Vectors 2019; 12:216. [PMID: 31064387 PMCID: PMC6505259 DOI: 10.1186/s13071-019-3471-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
Background Humans are susceptible to over 1400 pathogens. Co-infection by multiple pathogens is common, and can result in a range of neutral, facilitative, or antagonistic interactions within the host. Soil-transmitted helminths (STH) are powerful immunomodulators, but evidence of the effect of STH infection on the direction and magnitude of concurrent enteric microparasite infections is mixed. Methods We collected fecal samples from 891 randomly selected children and adults in rural Laos. Samples were analyzed for 5 STH species, 6 viruses, 9 bacteria, and 5 protozoa using a quantitative reverse transcription polymerase chain reaction (qRT-PCR) assay. We utilized logistic regression, controlling for demographics and household water, sanitation, and hygiene access, to examine the effect of STH infection on concurrent viral, bacterial, and protozoal infection. Results We found that STH infection was associated with lower odds of concurrent viral infection [odds ratio (OR): 0.48, 95% confidence interval (CI): 0.28–0.83], but higher odds of concurrent bacterial infections (OR: 1.81, 95% CI: 1.06–3.07) and concurrent protozoal infections (OR: 1.50, 95% CI: 0.95–2.37). Trends were consistent across STH species. Conclusions The impact of STH on odds of concurrent microparasite co-infection may differ by microparasite taxa, whereby STH infection was negatively associated with viral infections but positively associated with bacterial and protozoal infections. Results suggest that efforts to reduce STH through preventive chemotherapy could have a spillover effect on microparasite infections, though the extent of this impact requires additional study. The associations between STH and concurrent microparasite infection may reflect a reverse effect due to the cross-sectional study design. Additional research is needed to elucidate the exact mechanism of the immunomodulatory effects of STH on concurrent enteric microparasite infection. Electronic supplementary material The online version of this article (10.1186/s13071-019-3471-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna N Chard
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, 30322, USA
| | - Kelly K Baker
- Department of Occupational and Environmental Health, College of Public Health, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Kevin Tsai
- Department of Occupational and Environmental Health, College of Public Health, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Karen Levy
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, 30322, USA
| | - Jeticia R Sistrunk
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, 30322, USA
| | - Howard H Chang
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, 30322, USA
| | - Matthew C Freeman
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, 30322, USA.
| |
Collapse
|
17
|
Ascaris Larval Infection and Lung Invasion Directly Induce Severe Allergic Airway Disease in Mice. Infect Immun 2018; 86:IAI.00533-18. [PMID: 30249744 DOI: 10.1128/iai.00533-18] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/13/2018] [Indexed: 11/20/2022] Open
Abstract
Ascaris lumbricoides (roundworm) is the most common helminth infection globally and a cause of lifelong morbidity that may include allergic airway disease, an asthma phenotype. We hypothesize that Ascaris larval migration through the lungs leads to persistent airway hyperresponsiveness (AHR) and type 2 inflammatory lung pathology despite resolution of infection that resembles allergic airway disease. Mice were infected with Ascaris by oral gavage. Lung AHR was measured by plethysmography and histopathology with hematoxylin and eosin (H&E) and periodic acid-Schiff (PAS) stains, and cytokine concentrations were measured by using Luminex Magpix. Ascaris-infected mice were compared to controls or mice with allergic airway disease induced by ovalbumin (OVA) sensitization and challenge (OVA/OVA). Ascaris-infected mice developed profound AHR starting at day 8 postinfection (p.i.), peaking at day 12 p.i. and persisting through day 21 p.i., despite resolution of infection, which was significantly increased compared to controls and OVA/OVA mice. Ascaris-infected mice had a robust type 2 cytokine response in both the bronchoalveolar lavage (BAL) fluid and lung tissue, similar to that of the OVA/OVA mice, including interleukin-4 (IL-4) (P < 0.01 and P < 0.01, respectively), IL-5 (P < 0.001 and P < 0.001), and IL-13 (P < 0.001 and P < 0.01), compared to controls. By histopathology, Ascaris-infected mice demonstrated early airway remodeling similar to, but more profound than, that in OVA/OVA mice. We found that Ascaris larval migration causes significant pulmonary damage, including AHR and type 2 inflammatory lung pathology that resembles an extreme form of allergic airway disease. Our findings indicate that ascariasis may be an important cause of allergic airway disease in regions of endemicity.
Collapse
|
18
|
Caraballo L, Acevedo N, Zakzuk J. Ascariasis as a model to study the helminth/allergy relationships. Parasite Immunol 2018; 41:e12595. [PMID: 30295330 DOI: 10.1111/pim.12595] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 10/02/2018] [Indexed: 12/18/2022]
Abstract
Ascariasis is the most frequent soil transmitted helminthiasis and, as well as other helminth infections, is expected to influence the clinical presentation of allergic diseases such as asthma. Indeed, several clinical and experimental works have shown an important impact either increasing or suppressing symptoms, and the same effects have been detected on the underlying immune responses. In this review we analyze the work on this field performed in Colombia, a Latin American tropical country, including aspects such as the molecular genetics of the IgE response to Ascaris; the allergenic activity of Ascaris IgE-binding molecular components and the immunological and clinical influences of ascariasis on asthma. The analysis allows us to conclude that the impact of ascariasis on the inception and evolution of allergic diseases such as asthma deserves more investigation, but advances have been made during the last years. The concurrent parasite-induced immunostimulatory and immunosuppressive effects during this helminthiasis do modify the natural history of asthma and some aspects of the practice of allergology in the tropics. Theoretically it can also influence the epidemiological trends of allergic diseases either by its absence or presence in different regions and countries.
Collapse
Affiliation(s)
- Luis Caraballo
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| | - Nathalie Acevedo
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| | - Josefina Zakzuk
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| |
Collapse
|
19
|
Cytokines, Antibodies, and Histopathological Profiles during Giardia Infection and Variant-Specific Surface Protein-Based Vaccination. Infect Immun 2018; 86:IAI.00773-17. [PMID: 29555679 DOI: 10.1128/iai.00773-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/10/2018] [Indexed: 12/13/2022] Open
Abstract
Giardiasis is one of the most common human intestinal diseases worldwide. Several experimental animal models have been used to evaluate Giardia infections, with gerbils (Meriones unguiculatus) being the most valuable model due to their high susceptibility to Giardia infection, abundant shedding of cysts, and pathophysiological alterations and signs of disease similar to those observed in humans. Here, we report cytokine and antibody profiles both during the course of Giardia infection in gerbils and after immunization with a novel oral vaccine comprising a mixture of purified variant-specific surface proteins (VSPs). Transcript levels of representative cytokines of different immune profiles as well as macro- and microtissue alterations were assessed in Peyer's patches, mesenteric lymph nodes, and spleens. During infection, cytokine responses showed a biphasic profile: an early induction of Th1 (gamma interferon [IFN-γ], interleukin-1β [IL-1β], IL-6, and tumor necrosis factor [TNF]), Th17 (IL-17), and Th2 (IL-4) cytokines, together with intestinal alterations typical of inflammation, followed by a shift toward a predominant Th2 (IL-5) response, likely associated with a counterregulatory mechanism. Conversely, immunization with an oral vaccine comprising the entire repertoire of VSPs specifically showed high levels of IL-17, IL-6, IL-4, and IL-5, without obvious signs of inflammation. Both immunized and infected animals developed local (intestinal secretory IgA [S-IgA]) and systemic (serum IgG) humoral immune responses against VSPs; however, only infected animals showed evident signs of giardiasis. This is the first comprehensive report of cytokine expression and anti-Giardia antibody production during infection and VSP vaccination in gerbils, a reliable model of the human disease.
Collapse
|
20
|
Symeonidou I, Gelasakis AI, Arsenopoulos K, Angelou A, Beugnet F, Papadopoulos E. Feline gastrointestinal parasitism in Greece: emergent zoonotic species and associated risk factors. Parasit Vectors 2018; 11:227. [PMID: 29618378 PMCID: PMC5885467 DOI: 10.1186/s13071-018-2812-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/25/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Feline gastrointestinal parasitism constitutes an issue of concern for veterinarians since parasites are widespread and affect animals' health and welfare. Furthermore, some of these pathogens have zoonotic potential. To provide detailed data on the current epizootiology of feline endoparasitism, a multicentric survey was conducted during 2016. METHODS Faeces from 1150 cats were collected from all regions of Greece and examined by sedimentation and flotation techniques. Possible risk factors including gender, age, ownership status, living conditions and co-infections with other parasites were assessed using binary regression models for each one of the most prevalent parasites. RESULTS The overall gastrointestinal parasite prevalence in cats was 50.7%. The study population consisted of cats of both sexes, different age groups, ownership status and living conditions. A total of 10 gastrointestinal parasitic species were detected and up to 5 different parasites were isolated in the same faecal sample. The most frequently identified parasites were Toxocara cati (n = 278; 24.2%), followed by Cystoisospora spp. (n = 189; 16.4%), Ancylostomatidae (n = 186; 16.2%), Aelurostrongylus abstrusus (n = 40; 3.5%), Giardia spp. (n = 26; 2.3%), Joyeuxiella pasqualei (n = 14; 1.2%), Capillaria aerophila (n = 8; 0.7%), Dipylidium caninum (n = 3; 0.2%), Toxascaris leonina (n = 2; 0.1%) and Troglostrongylus brevior (n = 2; 0.1%). The occurrence of co-infections was 11.6%. Concerning risk factors, the likelihood of T. cati infection was higher for female cats living outdoors and for cats being infected with Cystoisospora spp. In the same frame, young, stray, male and free of A. abstrusus cats were more likely to be infected with Cystoisospora spp. Correspondingly, stray, infected with Giardia spp. but free of Cystoisospora spp. cats were more likely to be infected with Ancylostomatidae. Regarding A. abstrusus infection, a higher probability was reported for cats living outdoors and for cats free of Cystoisospora spp., while Giardia spp. infections were more common in young and co-infected with Ancylostomatidae animals. CONCLUSIONS The prevalence of parasitized cats in Greece was high and thus consideration should be paid to control the risk factors, to implement targeted preventive antiparasitic treatments and educate cat owners on the value of prevention for the health and welfare of their cats.
Collapse
Affiliation(s)
- Isaia Symeonidou
- Laboratory of Parasitology and Parasitic Diseases, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, PO Box: 393, GR 54124, Thessaloniki, Greece
| | - Athanasios I Gelasakis
- Veterinary Research Institute of Thessaloniki, ELGO-Demeter, GR 57001, Thermi, Thessaloniki, Greece
| | - Konstantinos Arsenopoulos
- Laboratory of Parasitology and Parasitic Diseases, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, PO Box: 393, GR 54124, Thessaloniki, Greece
| | - Athanasios Angelou
- Laboratory of Parasitology and Parasitic Diseases, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, PO Box: 393, GR 54124, Thessaloniki, Greece
| | - Frederic Beugnet
- Boehringer Ingelheim, 29 Avenue Tony Garnier, 69007, Lyon, France
| | - Elias Papadopoulos
- Laboratory of Parasitology and Parasitic Diseases, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, PO Box: 393, GR 54124, Thessaloniki, Greece.
| |
Collapse
|
21
|
Midttun HLE, Acevedo N, Skallerup P, Almeida S, Skovgaard K, Andresen L, Skov S, Caraballo L, van Die I, Jørgensen CB, Fredholm M, Thamsborg SM, Nejsum P, Williams AR. Ascaris Suum Infection Downregulates Inflammatory Pathways in the Pig Intestine In Vivo and in Human Dendritic Cells In Vitro. J Infect Dis 2017; 217:310-319. [DOI: 10.1093/infdis/jix585] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/07/2017] [Indexed: 11/15/2022] Open
|
22
|
Symeonidou I, Gelasakis AΙ, Arsenopoulos KV, Schaper R, Papadopoulos E. Regression models to assess the risk factors of canine gastrointestinal parasitism. Vet Parasitol 2017; 248:54-61. [PMID: 29173542 DOI: 10.1016/j.vetpar.2017.10.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/19/2017] [Accepted: 10/30/2017] [Indexed: 10/18/2022]
Abstract
Gastrointestinal parasites, protozoa and helminths, remain a common important finding in dogs presented to veterinary medical practices. Moreover, dogs may play an active role in the transmission of enteric parasites to humans, given their cohabitation, making canine gastrointestinal parasitism an issue of major concern for public health. To get an overview of the current state of canine gastrointestinal parasites' prevalence in Greece, a total of 1036 faecal samples were collected from dogs either presented in veterinary clinics or in animal shelters. Samples were examined by a combined sedimentation-flotation technique. Possible risk factors (age, ownership status, co-existence with other animals, coinfection with other parasites) were assessed using binary regression models for each one of the most prevalent parasites. The overall gastrointestinal parasite prevalence in dogs' faecal samples was 39%. A total of 11 intestinal parasitic species were detected and up to five different parasites were isolated in the same faecal sample. Toxocara canis was the most prevalent parasite in the studied population, followed by Isospora spp., Giardia spp., Uncinaria spp., Trichuris vulpis, Ancylostoma spp., Toxascaris leonina, and Dipylidium caninum. Additionally, other genera (Taenia, Capillaria and Angiostrongylus) were also detected in very low percentages. Young, stray, living without other dogs, coinfected with T. vulpis or T. canis and free of Isospora spp. dogs were more likely to be infected by Giardia spp. Likewise, young, stray, coinfected with T. vulpis or T. canis but free of Giardia spp. dogs were more likely to be infected by Isospora spp. T. canis infections were more common in dogs coinfected with Isospora spp., Giardia spp., D. caninum, Τaenia spp., T. vulpis and T. leonina Finally, T. vulpis more often infected older dogs and dogs coinfected with Isospora spp., Giardia spp. and T. canis Consequently, the prevalence of parasitized dogs was high and the diversity of parasites found was notable, which calls for a greater awareness among veterinarians and pet owners. The proposed prediction models should be taken under consideration in diagnostic approach of clinical cases, as well as in planning sustainable antiparasitic strategies.
Collapse
Affiliation(s)
- I Symeonidou
- Laboratory of Parasitology and Parasitic Diseases, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, PO Box: 393, GR 54124, Thessaloniki, Greece
| | - A Ι Gelasakis
- Veterinary Research Institute of Thessaloniki, ELGO-Demeter, GR 57001 Thermi, Thessaloniki, Greece
| | - K V Arsenopoulos
- Laboratory of Parasitology and Parasitic Diseases, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, PO Box: 393, GR 54124, Thessaloniki, Greece
| | - R Schaper
- Bayer Health Care AG, Animal Health, Leverkusen, Germany
| | - E Papadopoulos
- Laboratory of Parasitology and Parasitic Diseases, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, PO Box: 393, GR 54124, Thessaloniki, Greece.
| |
Collapse
|
23
|
Weatherhead J, Cortés AA, Sandoval C, Vaca M, Chico M, Loor S, Cooper PJ, Mejia R. Comparison of Cytokine Responses in Ecuadorian Children Infected with Giardia, Ascaris, or Both Parasites. Am J Trop Med Hyg 2017; 96:1394-1399. [PMID: 28719267 DOI: 10.4269/ajtmh.16-0580] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
AbstractMore than 2 billion people are infected with parasites globally, and the majority have coinfections. Intestinal protozoa and helminths induce polarizing CD4+ T-helper cell 1 (Th1) mediated cytokine responses within the host. Such immune polarization may inhibit the ability of the host to mount an adequate immune response for pathogen clearance to concurrent pathogens. The current study evaluated the plasma cytokine profile in Ascaris and Giardia coinfected children compared with Giardia- and Ascaris-only infected children. Fecal samples and blood samples were collected from asymptomatic 3-year-old children living in the district of Quininde, Ecuador. Stool samples that tested positive for Giardia lamblia-only, Ascaris lumbricoides-only, or G. lamblia and A. lumbricoides coinfections were confirmed by quantitative real-time polymerase chain reaction. Plasma samples from the study subjects were used to quantitate cytokines. A total of 39 patients were evaluated. Children with coinfection had a significant decrease in Th1 cytokine production, interleukin 2 (IL-2) (P < 0.05), IL-12 (P < 0.05), and tumor necrosis factor alpha (P < 0.05) compared with Giardia-only infected children. Coinfected children had an increase in IL-10/interferon gamma (IFN-γ) ratio compared with uninfected (P < 0.05) and Ascaris alone (P < 0.05). The increased IL-10/IFN-γ ratio in the setting of decreased Th1 cytokine response indicates Th2 polarization in the coinfected group. Reduced Th1 cytokines in children coinfected with Ascaris and Giardia may impair the host's ability to eradicate Giardia infection leading to chronic giardiasis.
Collapse
Affiliation(s)
- Jill Weatherhead
- National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas
| | | | - Carlos Sandoval
- Fundación Ecuatoriana para la Investigación en Salud, Quito, Ecuador
| | - Maritza Vaca
- Fundación Ecuatoriana para la Investigación en Salud, Quito, Ecuador
| | - Martha Chico
- Fundación Ecuatoriana para la Investigación en Salud, Quito, Ecuador
| | - Sophia Loor
- Fundación Ecuatoriana para la Investigación en Salud, Quito, Ecuador
| | - Philip J Cooper
- Institute of Infection and Immunity, St. George's University of London, London, United Kingdom.,Facultad de Ciencias Medicas, de la Salud y la Vida, Universidad Internacional del Ecuador, Quito, Ecuador.,Fundación Ecuatoriana para la Investigación en Salud, Quito, Ecuador
| | - Rojelio Mejia
- National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
24
|
Manko A, Motta JP, Cotton JA, Feener T, Oyeyemi A, Vallance BA, Wallace JL, Buret AG. Giardia co-infection promotes the secretion of antimicrobial peptides beta-defensin 2 and trefoil factor 3 and attenuates attaching and effacing bacteria-induced intestinal disease. PLoS One 2017. [PMID: 28622393 PMCID: PMC5473565 DOI: 10.1371/journal.pone.0178647] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Our understanding of polymicrobial gastrointestinal infections and their effects on host biology remains incompletely understood. Giardia duodenalis is an ubiquitous intestinal protozoan parasite infecting animals and humans. Concomitant infections with Giardia and other gastrointestinal pathogens commonly occur. In countries with poor sanitation, Giardia infection has been associated with decreased incidence of diarrheal disease and fever, and reduced serum inflammatory markers release, via mechanisms that remain obscure. This study analyzed Giardia spp. co-infections with attaching and effacing (A/E) pathogens, and assessed whether and how the presence of Giardia modulates host responses to A/E enteropathogens, and alters intestinal disease outcome. In mice infected with the A/E pathogen Citrobacter rodentium, co-infection with Giardia muris significantly attenuated weight loss, macro- and microscopic signs of colitis, bacterial colonization and translocation, while concurrently enhancing the production and secretion of antimicrobial peptides (AMPs) mouse β-defensin 3 and trefoil factor 3 (TFF3). Co-infection of human intestinal epithelial cells (Caco-2) monolayers with G. duodenalis trophozoites and enteropathogenic Escherichia coli (EPEC) enhanced the production of the AMPs human β-defensin 2 (HBD-2) and TFF3; this effect was inhibited with treatment of G. duodenalis with cysteine protease inhibitors. Collectively, these results suggest that Giardia infections are capable of reducing enteropathogen-induced colitis while increasing production of host AMPs. Additional studies also demonstrated that Giardia was able to directly inhibit the growth of pathogenic bacteria. These results reveal novel mechanisms whereby Giardia may protect against gastrointestinal disease induced by a co-infecting A/E enteropathogen. Our findings shed new light on how microbial-microbial interactions in the gut may protect a host during concomitant infections.
Collapse
Affiliation(s)
- Anna Manko
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, Alberta, Canada
| | - Jean-Paul Motta
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, Alberta, Canada
| | - James A. Cotton
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, Alberta, Canada
| | - Troy Feener
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
| | - Ayodele Oyeyemi
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, Alberta, Canada
| | - Bruce A. Vallance
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - John L. Wallace
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology & Pharmacology, University of Calgary, Alberta, Canada
| | - Andre G. Buret
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
25
|
González-Fernández D, Pons EDC, Rueda D, Sinisterra OT, Murillo E, Scott ME, Koski KG. C-reactive protein is differentially modulated by co-existing infections, vitamin deficiencies and maternal factors in pregnant and lactating indigenous Panamanian women. Infect Dis Poverty 2017; 6:94. [PMID: 28571565 PMCID: PMC5455098 DOI: 10.1186/s40249-017-0307-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 04/17/2017] [Indexed: 01/09/2023] Open
Abstract
Background The usefulness of C-reactive protein (CRP) as a non-specific marker of inflammation during pregnancy and lactation is unclear in impoverished populations where co-existing infections and vitamin deficiencies are common. Methods This cross-sectional study in Panama recruited 120 pregnant and 99 lactating Ngäbe-Buglé women from 14 communities in rural Panama. Obstetric history, indoor wood smoke exposure, fieldwork, BMI, vitamins A, B12, D, and folic acid, and inflammation markers (CRP, neutrophil/lymphocyte ratio (NLR), plateletcrit and cytokines) were measured. Multiple regressions explored both associations of CRP with other inflammatory markers and associations of CRP and elevated CRP based on trimester-specific cut-offs with maternal factors, infections and vitamin deficiencies. Results CRP was higher in pregnancy (51.4 ± 4.7 nmol/L) than lactation (27.8 ± 3.5 nmol/L) and was elevated above trimester specific cut-offs in 21% of pregnant and 30% of lactating women. Vitamin deficiencies were common (vitamin A 29.6%; vitamin D 68.5%; vitamin B12 68%; folic acid 25.5%) and over 50% of women had two or more concurrent deficiencies as well as multiple infections. Multiple regression models highlighted differences in variables associated with CRP between pregnancy and lactation. In pregnancy, CRP was positively associated with greater indoor wood smoke exposure, caries and hookworm and negatively associated with Ascaris and vaginal Lactobacillus and Bacteroides/Gardnerella scores. Consistent with this, greater wood smoke exposure, caries as well as higher diplococcal infection score increased the odds of trimester-elevated CRP concentrations whereas longer gestational age lowered the likelihood of a trimester-elevated CRP. During lactation, folic acid deficiency was associated with higher CRP whereas parity, number of eosinophils and Mobiluncus score were associated with lower CRP. Also, a higher BMI and Trichomonas vaginalis score increased the likelihood of an elevated CRP whereas higher parity and number of eosinophils were associated with lower likelihood of an elevated CRP. Conclusions Infections both raise and lower CRP concentrations in pregnant and lactating mothers. Only folic acid deficiency during lactation was associated with higher CRP concentrations. Caution is required when interpreting CRP concentrations in pregnant and lactating women who have co-existing nutrient deficiencies and multiple infections. Electronic supplementary material The online version of this article (doi:10.1186/s40249-017-0307-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Doris González-Fernández
- School of Dietetics and Human Nutrition, Macdonald Campus of McGill University, 21,111 Lakeshore Road, Ste-Anne-de-Bellevue, QC, H9X 3V9, Canada
| | | | - Delfina Rueda
- "Comarca Ngäbe-Buglé" Health Region, Ministry of Health, San Félix, Chiriquí Province, Panama
| | - Odalis Teresa Sinisterra
- "Panamá Norte" Health Region, Ministry of Health, Las Cumbres Square, Transithmian Highway, Panama City, Panama
| | - Enrique Murillo
- Department of Biochemistry, University of Panama, Simón Bolivar Avenue (Transithmian Highway), Panama City, Panama.,Department of Biochemistry, University of Panama, Manuel Espinoza Batista and Jose De Fabrega Avenues, Panama City, Panama
| | - Marilyn E Scott
- Institute of Parasitology and Centre for Host-Parasite Interactions, Macdonald Campus of McGill University, 21,111 Lakeshore Road, Ste-Anne-de-Bellevue, QC, H9X 3 V9, Canada
| | - Kristine G Koski
- School of Dietetics and Human Nutrition and Centre for Host-Parasite Interactions, Macdonald Campus, McGill University, 21,111 Lakeshore Road, Ste-Anne-de-Bellevue, QC, H9X 3 V9, Canada.
| |
Collapse
|
26
|
Pérez-Chacón G, Pocaterra LA, Rojas E, Hernán A, Jiménez JC, Núñez L. Coinfection with Hymenolepis nana, Hymenolepis diminuta, Giardia intestinalis, and Human Immunodeficiency Virus: A Case Report with Complex Immunologic Interactions. Am J Trop Med Hyg 2017; 96:1094-1096. [PMID: 28219994 DOI: 10.4269/ajtmh.16-0413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AbstractWe describe the case of a 43-year-old human immunodeficiency virus-infected man receiving combined antiretroviral therapy and coinfected with Hymenolepis nana, Hymenolepis diminuta, and Giardia intestinalis, presenting as chronic diarrhea and critical weight loss. Immunological aspects of these interactions are reviewed.
Collapse
Affiliation(s)
- Gladymar Pérez-Chacón
- Cátedra de Parasitología, Escuela de Medicina "José María Vargas," Facultad de Medicina de la Universidad Central de Venezuela, Caracas, Venezuela
| | - Leonor A Pocaterra
- Cátedra de Parasitología, Escuela de Medicina "José María Vargas," Facultad de Medicina de la Universidad Central de Venezuela, Caracas, Venezuela
| | - Elsy Rojas
- Laboratorio de Parasitosis Intestinales, Escuela de Medicina "José María Vargas," Facultad de Medicina de la Universidad Central de Venezuela, Caracas, Venezuela
| | - Aurora Hernán
- Cátedra de Parasitología, Escuela de Medicina "José María Vargas," Facultad de Medicina de la Universidad Central de Venezuela, Caracas, Venezuela
| | - Juan Carlos Jiménez
- Laboratorio de Bioquímica, Instituto de Inmunología, Facultad de Medicina de la Universidad Central de Venezuela, Caracas, Venezuela
| | - Luz Núñez
- Cátedra de Parasitología, Escuela de Medicina "José María Vargas," Facultad de Medicina de la Universidad Central de Venezuela, Caracas, Venezuela
| |
Collapse
|
27
|
Allain T, Amat CB, Motta JP, Manko A, Buret AG. Interactions of Giardia sp. with the intestinal barrier: Epithelium, mucus, and microbiota. Tissue Barriers 2017; 5:e1274354. [PMID: 28452685 DOI: 10.1080/21688370.2016.1274354] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Understanding how intestinal enteropathogens cause acute and chronic alterations has direct animal and human health perspectives. Significant advances have been made on this field by studies focusing on the dynamic crosstalk between the intestinal protozoan parasite model Giardia duodenalis and the host intestinal mucosa. The concept of intestinal barrier function is of the highest importance in the context of many gastrointestinal diseases such as infectious enteritis, inflammatory bowel disease, and post-infectious gastrointestinal disorders. This crucial function relies on 3 biotic and abiotic components, first the commensal microbiota organized as a biofilm, then an overlaying mucus layer, and finally the tightly structured intestinal epithelium. Herein we review multiple strategies used by Giardia parasite to circumvent these 3 components. We will summarize what is known and discuss preliminary observations suggesting how such enteropathogen directly and/ or indirectly impairs commensal microbiota biofilm architecture, disrupts mucus layer and damages host epithelium physiology and survival.
Collapse
Affiliation(s)
- Thibault Allain
- a Department of Biological Sciences , University of Calgary , Calgary , AB , Canada.,b Inflammation Research Network, University of Calgary , Calgary , AB , Canada.,c Host-Parasite Interactions, University of Calgary , Calgary , AB , Canada
| | - Christina B Amat
- a Department of Biological Sciences , University of Calgary , Calgary , AB , Canada.,b Inflammation Research Network, University of Calgary , Calgary , AB , Canada.,c Host-Parasite Interactions, University of Calgary , Calgary , AB , Canada
| | - Jean-Paul Motta
- a Department of Biological Sciences , University of Calgary , Calgary , AB , Canada.,b Inflammation Research Network, University of Calgary , Calgary , AB , Canada.,c Host-Parasite Interactions, University of Calgary , Calgary , AB , Canada
| | - Anna Manko
- a Department of Biological Sciences , University of Calgary , Calgary , AB , Canada.,b Inflammation Research Network, University of Calgary , Calgary , AB , Canada.,c Host-Parasite Interactions, University of Calgary , Calgary , AB , Canada
| | - André G Buret
- a Department of Biological Sciences , University of Calgary , Calgary , AB , Canada.,b Inflammation Research Network, University of Calgary , Calgary , AB , Canada.,c Host-Parasite Interactions, University of Calgary , Calgary , AB , Canada
| |
Collapse
|
28
|
Concomitant helminth infection downmodulates the Vaccinia virus-specific immune response and potentiates virus-associated pathology. Int J Parasitol 2016; 47:1-10. [PMID: 28003150 DOI: 10.1016/j.ijpara.2016.08.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/29/2016] [Accepted: 08/31/2016] [Indexed: 12/16/2022]
Abstract
The aim of this work was to elucidate the immunopathological mechanisms of how helminths may influence the course of a viral infection, using a murine model. Severe virulence, a relevant increase in the virus titres in the lung and a higher mortality rate were observed in Ascaris and Vaccinia virus (VACV) co-infected mice, compared with VACV mono-infected mice. Immunopathological analysis suggested that the ablation of CD8+ T cells, the marked reduction of circulating CD4+ T cells producing IFN-γ, and the robust pulmonary inflammation were associated with the increase of morbidity/mortality in co-infection and subsequently with the negative impact of concomitant pulmonary ascariasis and respiratory VACV infection for the host. On the other hand, when evaluating the impact of the co-infection on the parasitic burden, co-infected mice presented a marked decrease in the total number of migrating Ascaris lung-stage larvae in comparison with Ascaris mono-infection. Taken together, our major findings suggest that Ascaris and VACV co-infection may potentiate the virus-associated pathology by the downmodulation of the VACV-specific immune response. Moreover, this study provides new evidence of how helminth parasites may influence the course of a coincident viral infection.
Collapse
|
29
|
Cotton JA, Amat CB, Buret AG. Disruptions of Host Immunity and Inflammation by Giardia Duodenalis: Potential Consequences for Co-Infections in the Gastro-Intestinal Tract. Pathogens 2015; 4:764-92. [PMID: 26569316 PMCID: PMC4693164 DOI: 10.3390/pathogens4040764] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/04/2015] [Accepted: 11/05/2015] [Indexed: 12/11/2022] Open
Abstract
Giardia duodenalis (syn. G. intestinalis, or G. lamblia) is a leading cause of waterborne diarrheal disease that infects hundreds of millions of people annually. Research on Giardia has greatly expanded within the last few years, and our understanding of the pathophysiology and immunology on this parasite is ever increasing. At peak infection, Giardia trophozoites induce pathophysiological responses that culminate in the development of diarrheal disease. However, human data has suggested that the intestinal mucosa of Giardia-infected individuals is devoid of signs of overt intestinal inflammation, an observation that is reproduced in animal models. Thus, our understanding of host inflammatory responses to the parasite remain incompletely understood and human studies and experimental data have produced conflicting results. It is now also apparent that certain Giardia infections contain mechanisms capable of modulating their host's immune responses. As the oral route of Giardia infection is shared with many other gastrointestinal (GI) pathogens, co-infections may often occur, especially in places with poor sanitation and/or improper treatment of drinking water. Moreover, Giardia infections may modulate host immune responses and have been found to protect against the development of diarrheal disease in developing countries. The following review summarizes our current understanding of the immunomodulatory mechanisms of Giardia infections and their consequences for the host, and highlights areas for future research. Potential implications of these immunomodulatory effects during GI co-infection are also discussed.
Collapse
Affiliation(s)
- James A Cotton
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
| | - Christina B Amat
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada.
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada.
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada.
| | - Andre G Buret
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada.
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada.
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada.
| |
Collapse
|
30
|
Abstract
The term “immune” pertains to the body keeping itself free from diseases, not to trigger any diseases. In this regard, it makes sense for us to divide antigenicity into immunogenicity and allergenicity. This distinction allows for the characterization of all types of modern antigens, i.e., to evaluate and modify a priori the allergenicity of an antigen before it is applied to humans. In this chapter, we also formulated the hypothesis that “Balanced Stimulation by Whole Antigens” is essential for immune development. This hypothesis revives the practicality of the “Hygiene Hypothesis” and can provide a fundamental solution to curb the increasing prevalence of allergic disease, namely, early exposure, at 0–1 year old or earlier, in utero, of representative allergens/protein antigens with immunogenicity retained or improved and allergenicity attenuated or eliminated.
Collapse
|
31
|
|
32
|
Cimino RO, Jeun R, Juarez M, Cajal PS, Vargas P, Echazú A, Bryan PE, Nasser J, Krolewiecki A, Mejia R. Identification of human intestinal parasites affecting an asymptomatic peri-urban Argentinian population using multi-parallel quantitative real-time polymerase chain reaction. Parasit Vectors 2015; 8:380. [PMID: 26183074 PMCID: PMC4504406 DOI: 10.1186/s13071-015-0994-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/07/2015] [Indexed: 11/29/2022] Open
Abstract
Background In resource-limited countries, stool microscopy is the diagnostic test of choice for intestinal parasites (soil-transmitted helminths and/or intestinal protozoa). However, sensitivity and specificity is low. Improved diagnosis of intestinal parasites is especially important for accurate measurements of prevalence and intensity of infections in endemic areas. Methods The study was carried out in Orán, Argentina. A total of 99 stool samples from a local surveillance campaign were analyzed by concentration microscopy and McMaster egg counting technique compared to the analysis by multi-parallel quantitative real-time polymerase chain reaction (qPCR). This study compared the performance of qPCR assay and stool microscopy for 8 common intestinal parasites that infect humans including the helminths Ascaris lumbricoides, Ancylostoma duodenale, Necator americanus, Strongyloides stercoralis, Trichuris trichiura, and the protozoa Giardia lamblia, Cryptosporidium parvum/hominis, and Entamoeba histolytica, and investigated the prevalence of polyparasitism in an endemic area. Results qPCR showed higher detection rates for all parasites as compared to stool microscopy except T. trichiura. Species-specific primers and probes were able to distinguish between A. duodenale (19.1 %) and N. americanus (36.4 %) infections. There were 48.6 % of subjects co-infected with both hookworms, and a significant increase in hookworm DNA for A. duodenale versus N. americanus (119.6 fg/μL: 0.63 fg/μL, P < 0.001) respectively. qPCR outperformed microscopy by the largest margin in G. lamblia infections (63.6 % versus 8.1 %, P < 0.05). Polyparasitism was detected more often by qPCR compared to microscopy (64.7 % versus 24.2 %, P < 0.05). Conclusions Multi-parallel qPCR is a quantitative molecular diagnostic method for common intestinal parasites in an endemic area that has improved diagnostic accuracy compared to stool microscopy. This first time use of multi-parallel qPCR in Argentina has demonstrated the high prevalence of intestinal parasites in a peri-urban area. These results will contribute to more accurate epidemiological survey, refined treatment strategies on a public scale, and better health outcomes in endemic settings.
Collapse
Affiliation(s)
- Rubén O Cimino
- Instituto de Investigaciones en Enfermedades Tropicales - Universidad Nacional de Salta/CONICET, Oran, Argentina. .,Cátedra de Química Biológica, Facultad de Ciencias Naturales, Universidad Nacional de Salta, Oran, Argentina.
| | - Rebecca Jeun
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Marisa Juarez
- Instituto de Investigaciones en Enfermedades Tropicales - Universidad Nacional de Salta/CONICET, Oran, Argentina. .,Fundación Mundo Sano, Buenos Aires, Argentina.
| | - Pamela S Cajal
- Instituto de Investigaciones en Enfermedades Tropicales - Universidad Nacional de Salta/CONICET, Oran, Argentina.
| | - Paola Vargas
- Instituto de Investigaciones en Enfermedades Tropicales - Universidad Nacional de Salta/CONICET, Oran, Argentina.
| | - Adriana Echazú
- Instituto de Investigaciones en Enfermedades Tropicales - Universidad Nacional de Salta/CONICET, Oran, Argentina.
| | - Patricia E Bryan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Julio Nasser
- Instituto de Investigaciones en Enfermedades Tropicales - Universidad Nacional de Salta/CONICET, Oran, Argentina. .,Cátedra de Química Biológica, Facultad de Ciencias Naturales, Universidad Nacional de Salta, Oran, Argentina.
| | - Alejandro Krolewiecki
- Instituto de Investigaciones en Enfermedades Tropicales - Universidad Nacional de Salta/CONICET, Oran, Argentina.
| | - Rojelio Mejia
- Cátedra de Química Biológica, Facultad de Ciencias Naturales, Universidad Nacional de Salta, Oran, Argentina. .,National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
33
|
Buret AG, Amat CB, Manko A, Beatty JK, Halliez MCM, Bhargava A, Motta JP, Cotton JA. Giardia duodenalis: New Research Developments in Pathophysiology, Pathogenesis, and Virulence Factors. CURRENT TROPICAL MEDICINE REPORTS 2015. [DOI: 10.1007/s40475-015-0049-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
34
|
Cotton JA, Motta JP, Schenck LP, Hirota SA, Beck PL, Buret AG. Giardia duodenalis infection reduces granulocyte infiltration in an in vivo model of bacterial toxin-induced colitis and attenuates inflammation in human intestinal tissue. PLoS One 2014; 9:e109087. [PMID: 25289678 PMCID: PMC4188619 DOI: 10.1371/journal.pone.0109087] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 09/08/2014] [Indexed: 12/22/2022] Open
Abstract
Giardia duodenalis (syn. G. intestinalis, G. lamblia) is a predominant cause of waterborne diarrheal disease that may lead to post-infectious functional gastrointestinal disorders. Although Giardia-infected individuals could carry as much as 106 trophozoites per centimetre of gut, their intestinal mucosa is devoid of overt signs of inflammation. Recent studies have shown that in endemic countries where bacterial infectious diseases are common, Giardia infections can protect against the development of diarrheal disease and fever. Conversely, separate observations have indicated Giardia infections may enhance the severity of diarrheal disease from a co-infecting pathogen. Polymorphonuclear leukocytes or neutrophils (PMNs) are granulocytic, innate immune cells characteristic of acute intestinal inflammatory responses against bacterial pathogens that contribute to the development of diarrheal disease following recruitment into intestinal tissues. Giardia cathepsin B cysteine proteases have been shown to attenuate PMN chemotaxis towards IL-8/CXCL8, suggesting Giardia targets PMN accumulation. However, the ability of Giardia infections to attenuate PMN accumulation in vivo and how in turn this effect may alter the host inflammatory response in the intestine has yet to be demonstrated. Herein, we report that Giardia infection attenuates granulocyte tissue infiltration induced by intra-rectal instillation of Clostridium difficile toxin A and B in an isolate-dependent manner. This attenuation of granulocyte infiltration into colonic tissues paralled decreased expression of several cytokines associated with the recruitment of PMNs. Giardia trophozoite isolates that attenuated granulocyte infiltration in vivo also decreased protein expression of cytokines released from inflamed mucosal biopsy tissues collected from patients with active Crohn’s disease, including several cytokines associated with PMN recruitment. These results demonstrate for the first time that certain Giardia infections may attenuate PMN accumulation by decreasing the expression of the mediators responsible for their recruitment.
Collapse
Affiliation(s)
- James A. Cotton
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, Alberta, Canada
| | - Jean-Paul Motta
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, Alberta, Canada
| | - L. Patrick Schenck
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Simon A. Hirota
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Department of Immunology, Microbiology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Paul L. Beck
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andre G. Buret
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
35
|
Comparative community-level associations of helminth infections and microparasite shedding in wild long-tailed macaques in Bali, Indonesia. Parasitology 2014; 142:480-9. [PMID: 25249163 DOI: 10.1017/s0031182014001462] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Helminthes have the capacity to modulate host immunity, leading to positive interactions with coinfecting microparasites. This phenomenon has been primarily studied during coinfections with a narrow range of geo-helminthes and intracellular microparasites in human populations or under laboratory conditions. Far less is known regarding differences in coinfection dynamics between helminth types, the range of microparasites that might be affected or the overall community-level effects of helminth infections on microparasites in wild systems. Here, we analysed the presence/absence and abundance patterns of enteric parasites in long-tailed macaques (Macaca fascicularis) on the island of Bali, Indonesia, to assess whether naturally occurring helminth infections were associated with increased shedding of the most common intracellular (Cryptosporidium spp., Isospora spp.) and extracellular (Entamoeba spp., Giardia spp.) microparasites. We also comparatively assessed the statistical correlations of different helminth taxa with microparasite shedding to determine if there were consistent relationships between the specific helminth taxa and microparasites. Helminth infections were associated with increased shedding of both intracellular and extracellular microparasites. Platyhelminthes repeatedly displayed strong positive correlations with several microparasites; while nematodes did not. Our results indicate that helminthes can influence microparasite community shedding dynamics under wild conditions, but that trends may be driven by a narrow range of helminthes.
Collapse
|
36
|
Escobedo AA, Hanevik K, Almirall P, Cimerman S, Alfonso M. Management of chronic Giardia infection. Expert Rev Anti Infect Ther 2014; 12:1143-57. [PMID: 25059638 DOI: 10.1586/14787210.2014.942283] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Advances in our understanding of chronic giardiasis (CG) may improve our care of patients in this stage of the disease. This review proposes a new concept of CG and highlights the recent advances in our understanding and management of this condition. According to this review, management requires, initially, an accurate diagnosis, which may exclude several conditions that can mimic CG. Optimal treatment requires a tailored approach which includes the recognition of the known modifiable causes of this health condition, assessment of symptoms and potential complications, their treatment utilizing, if necessary, a multidisciplinary team, and an ongoing monitoring for the effect of therapy - weighing the efficacy of individual drugs - all of these together may lead to a successful treatment of CG.
Collapse
Affiliation(s)
- Angel A Escobedo
- Academic Paediatric Hospital "Pedro Borrás", Calle F No. 616 esquina 27, Plaza, La Habana, CP 10400, Cuba
| | | | | | | | | |
Collapse
|
37
|
Tan X, Li D, Wang X, Zeng Y, Yan Y, Yang L. Claudin-2 downregulation by KSHV infection is involved in the regulation of endothelial barrier function. J Cutan Pathol 2014; 41:630-9. [PMID: 24995964 DOI: 10.1111/cup.12332] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/01/2014] [Accepted: 02/17/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND Kaposi sarcoma (KS), caused by the infection of Kaposi sarcoma-associated herpesvirus (KSHV), is a disease manifested mainly by dark purple skin and mouth nodules. Cancer care studies showed that co-infection of KSHV and human immunodeficiency virus (HIV) was able to increase the patients' survival, but the underlying mechanisms are still elusive. METHODS To understand the mechanism underlying the prolonged survival in KSHV-HIV co-infected patients, we performed microarray analysis on RNA extracted from biopsies from KS tumors and adjacent healthy tissues in four KS patients. Subsequently, we performed hierarchical clustering, gene ontology (GO) and ingenuity pathway analysis. We then characterized the roles of tight junction protein claudin-2 in the endothelial barrier function. RESULTS Three hundred and forty-three differentially expressed genes were identified, of which 246 genes exhibited significantly increased expression in the tumor compared to the adjacent healthy tissue and 97 genes showed downregulated expression, including claudin-2. Knockdown of claudin-2 in cultured endothelial cells enhances barrier function by altering the charge selectivity, but not the size selectivity. CONCLUSION Claudin-2 expression is decreased in KS tumors from patients co-infected with KSHV and HIV. Decreased claudin-2 enhances endothelial barrier function and may play a role in the prolonged survival of patients with KSHV and HIV co-infection.
Collapse
Affiliation(s)
- Xiaohua Tan
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | | | | | | | | | | |
Collapse
|
38
|
Wanyiri JW, Kanyi H, Maina S, Wang DE, Steen A, Ngugi P, Kamau T, Waithera T, O'Connor R, Gachuhi K, Wamae CN, Mwamburi M, Ward HD. Cryptosporidiosis in HIV/AIDS patients in Kenya: clinical features, epidemiology, molecular characterization and antibody responses. Am J Trop Med Hyg 2014; 91:319-28. [PMID: 24865675 DOI: 10.4269/ajtmh.13-0254] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We investigated the epidemiological and clinical features of cryptosporidiosis, the molecular characteristics of infecting species and serum antibody responses to three Cryptosporidium-specific antigens in human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome (AIDS) patients in Kenya. Cryptosporidium was the most prevalent enteric pathogen and was identified in 56 of 164 (34%) of HIV/AIDS patients, including 25 of 70 (36%) with diarrhea and 31 of 94 (33%) without diarrhea. Diarrhea in patients exclusively infected with Cryptosporidium was significantly associated with the number of children per household, contact with animals, and water treatment. Cryptosporidium hominis was the most prevalent species and the most prevalent subtype family was Ib. Patients without diarrhea had significantly higher serum IgG levels to Chgp15, Chgp40 and Cp23, and higher fecal IgA levels to Chgp15 and Chgp40 than those with diarrhea suggesting that antibody responses to these antigens may be associated with protection from diarrhea and supporting further investigation of these antigens as vaccine candidates.
Collapse
Affiliation(s)
- Jane W Wanyiri
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| | - Henry Kanyi
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| | - Samuel Maina
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| | - David E Wang
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| | - Aaron Steen
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| | - Paul Ngugi
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| | - Timothy Kamau
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| | - Tabitha Waithera
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| | - Roberta O'Connor
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| | - Kimani Gachuhi
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| | - Claire N Wamae
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| | - Mkaya Mwamburi
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| | - Honorine D Ward
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts; Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts; Center of Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya; Center of Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya; Kenyatta National Hospital, Nairobi, Kenya
| |
Collapse
|
39
|
Hagel I, Puccio F, López E, Lugo D, Cabrera M, Di Prisco MC. Intestinal parasitic infections and atopic dermatitis among Venezuelan Warao Amerindian pre- school children. Pediatr Allergy Immunol 2014; 25:276-82. [PMID: 24417507 DOI: 10.1111/pai.12190] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND We evaluated the influence of intestinal parasitic infection on food sensitization associated to the severity of Atopic Dermatitis (AD) in a group of Warao Amerindian pre- school children. METHODS Feces examinations were performed in fresh stool specimens. Diagnosis of AD was done according to Hannifin and Rajka criteria and SCORAD index. Skin prick tests (SPT) were performed using extracts of cow's milk (CM), hen's egg (HE), Dermatophagoides pteronyssinus and Dermatophagoides farinae . Serum CM and HE-IgE levels (ELISA) were measured. Quantikine (R&D systems) assays were used for the determination of IL-13, TNF-α IL-6, and sCD23 in supernatants of CM- and HE- whole blood stimulated samples. RESULTS Atopic Dermatitis was reported in 23% of the children. It was significantly (p < 0.0001) associated toward both CM and HE- SPT positivity (p < 0.001). Giardia duodenalis infection (37%) was associated to the presence of AD (p = 0.005) and to a significant increase in the levels of CM stimulated TNF-α (p=0.006), IL-13 (p = 0.01), sCD23 (p = 0.001), CM-IgE (p = 0.012) and CM-SPT (p < 0.0001). Similarly, G. duodenalis infection was particularly associated with the increase on the levels of HE-stimulated TNF-α (p = 0.001), sCD23 (p = 0.001), HE-IgE levels (p = 0.002) and HE-SPT (p = 0.001). CONCLUSION Gut inflammation caused by G. duodenalis may enhance food allergic reactivity contributing to the manifestation of AD in these children. However, other environmental factors (not considered in this work) as well as an atopic background among the Warao population would also contribute to the presence of AD.
Collapse
Affiliation(s)
- Isabel Hagel
- Institute of Biomedicine, Faculty of Medicine, Central University of Venezuela, Caracas, Venezuela
| | | | | | | | | | | |
Collapse
|
40
|
Blackwell AD, Martin M, Kaplan H, Gurven M. Antagonism between two intestinal parasites in humans: the importance of co-infection for infection risk and recovery dynamics. Proc Biol Sci 2013; 280:20131671. [PMID: 23986108 DOI: 10.1098/rspb.2013.1671] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Co-infection may affect transmission and recovery from infection, but remains an understudied element of disease ecology, particularly with regard to antagonism between parasites sharing a host. Helminth and giardia infections are often endemic in the same populations and both occupy the small intestine; yet few studies have examined interactions between these parasites. We report on helminth-giardia co-infections in a panel study of forager-horticulturalists in the Bolivian lowlands. Parasites were identified in faecal samples from 3275 participants, collected during 5235 medical exams over 6 years. Longitudinal co-infection patterns were examined using logistic mixed and multi-state Markov models. The most prevalent infections were hookworm (56%), Giardia lamblia (30%) and Ascaris lumbricoides (15%). Cross-sectionally, hookworm and A. lumbricoides were negatively associated with G. lamblia (OR = 0.60; OR = 0.65, respectively). Longitudinally, giardia infection was less likely in helminth-infected individuals (HR: 0.46). Infection with helminths was also less likely for individuals infected with giardia (HR: 0.71). Finally, treatment with mebendazole reduced subsequent hookworm infections, but resulted in a marginal increase in the odds of G. lamblia infection. Our results provide evidence for an antagonistic relationship between helminths and giardia, and suggest that co-infection should be considered in disease transmission models and treatment decisions.
Collapse
Affiliation(s)
- Aaron D Blackwell
- Department of Anthropology, University of California, Santa Barbara, CA 93106-3210, USA.
| | | | | | | |
Collapse
|
41
|
Bartelt LA, Roche J, Kolling G, Bolick D, Noronha F, Naylor C, Hoffman P, Warren C, Singer S, Guerrant R. Persistent G. lamblia impairs growth in a murine malnutrition model. J Clin Invest 2013; 123:2672-84. [PMID: 23728173 PMCID: PMC3668820 DOI: 10.1172/jci67294] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 03/15/2013] [Indexed: 12/14/2022] Open
Abstract
Giardia lamblia infections are nearly universal among children in low-income countries and are syndemic with the triumvirate of malnutrition, diarrhea, and developmental growth delays. Amidst the morass of early childhood enteropathogen exposures in these populations, G. lamblia–specific associations with persistent diarrhea, cognitive deficits, stunting, and nutrient deficiencies have demonstrated conflicting results, placing endemic pediatric giardiasis in a state of equipoise. Many infections in endemic settings appear to be asymptomatic/ subclinical, further contributing to uncertainty regarding a causal link between G. lamblia infection and developmental delay. We used G. lamblia H3 cyst infection in a weaned mouse model of malnutrition to demonstrate that persistent giardiasis leads to epithelial cell apoptosis and crypt hyperplasia. Infection was associated with a Th2-biased inflammatory response and impaired growth. Malnutrition accentuated the severity of these growth decrements. Faltering malnourished mice exhibited impaired compensatory responses following infection and demonstrated an absence of crypt hyperplasia and subsequently blunted villus architecture. Concomitantly, severe malnutrition prevented increases in B220+ cells in the lamina propria as well as mucosal Il4 and Il5 mRNA in response to infection. These findings add insight into the potential role of G. lamblia as a "stunting" pathogen and suggest that, similarly, malnourished children may be at increased risk of G. lamblia– potentiated growth decrements.
Collapse
Affiliation(s)
- Luther A Bartelt
- Division of Infectious Diseases and Center for Global Health, University of Virginia, Charlottesville, Virginia 22903, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abanyie FA, McCracken C, Kirwan P, Molloy SF, Asaolu SO, Holland CV, Gutman J, Lamb TJ. Ascaris co-infection does not alter malaria-induced anaemia in a cohort of Nigerian preschool children. Malar J 2013; 12:1. [PMID: 23282136 PMCID: PMC3544581 DOI: 10.1186/1475-2875-12-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 12/26/2012] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Co-infection with malaria and intestinal parasites such as Ascaris lumbricoides is common. Malaria parasites induce a pro-inflammatory immune response that contributes to the pathogenic sequelae, such as malarial anaemia, that occur in malaria infection. Ascaris is known to create an anti-inflammatory immune environment which could, in theory, counteract the anti-malarial inflammatory immune response, minimizing the severity of malarial anaemia. This study examined whether Ascaris co-infection can minimize the severity of malarial anaemia. METHODS Data from a randomized controlled trial on the effect of antihelminthic treatment in Nigerian preschool-aged (6-59 months) children conducted in 2006-2007 were analysed to examine the effect of malaria and Ascaris co-infection on anaemia severity. Children were enrolled and tested for malaria, helminths and anaemia at baseline, four, and eight months. Six hundred and ninety subjects were analysed in this study. Generalized linear mixed models were used to assess the relationship between infection status and Ascaris and Plasmodium parasite intensity on severity of anaemia, defined as a haemoglobin less than 11 g/dL. RESULTS Malaria prevalence ranged from 35-78% over the course of this study. Of the malaria-infected children, 55% were co-infected with Ascaris at baseline, 60% were co-infected four months later and 48% were co-infected eight months later, underlining the persistent prevalence of malaria-nematode co-infections in this population. Over the course of the study the percentage of anaemic subjects in the population ranged between 84% at baseline and 77% at the eight-month time point. The odds of being anaemic were four to five times higher in children infected with malaria compared to those without malaria. Ascaris infection alone did not increase the odds of being anaemic, indicating that malaria was the main cause of anaemia in this population. There was no significant difference in the severity of anaemia between children singly infected with malaria and co-infected with malaria and Ascaris. CONCLUSION In this cohort of Nigerian preschool children, malaria infection was the major contributor to anaemia status. Ascaris co-infection neither exacerbated nor ameliorated the severity of malarial anaemia.
Collapse
|