Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Brain Commun ; 5(5): fcad223, 2023.
Article in English | MEDLINE | ID: mdl-37731906

ABSTRACT

Nodding syndrome is a neglected, disabling and potentially fatal epileptic disorder of unknown aetiology affecting thousands of individuals mostly confined to Eastern sub-Saharan Africa. Previous studies have identified multiple associations-including Onchocerca volvulus, antileiomodin-1 antibodies, vitamin B6 deficiency and measles virus infection-yet, none is proven causal. We conducted a case-control study of children with early-stage nodding syndrome (symptom onset <1 year). Cases and controls were identified through a household survey in the Greater Mundri area in South Sudan. A wide range of parasitic, bacterial, viral, immune-mediated, metabolic and nutritional risk factors was investigated using conventional and state-of-the-art untargeted assays. Associations were examined by multiple logistic regression analysis, and a hypothetical causal model was constructed using structural equation modelling. Of 607 children with nodding syndrome, 72 with early-stage disease were included as cases and matched to 65 household- and 44 community controls. Mansonella perstans infection (odds ratio 7.04, 95% confidence interval 2.28-21.7), Necator americanus infection (odds ratio 2.33, 95% confidence interval 1.02-5.3), higher antimalarial seroreactivity (odds ratio 1.75, 95% confidence interval 1.20-2.57), higher vitamin E concentration (odds ratio 1.53 per standard deviation increase, 95% confidence interval 1.07-2.19) and lower vitamin B12 concentration (odds ratio 0.56 per standard deviation increase, 95% confidence interval 0.36-0.87) were associated with higher odds of nodding syndrome. In a structural equation model, we hypothesized that Mansonella perstans infection, higher vitamin E concentration and fewer viral exposures increased the risk of nodding syndrome while lower vitamin B12 concentration, Necator americanus and malaria infections resulted from having nodding syndrome. We found no evidence that Onchocerca volvulus, antileiomodin-1 antibodies, vitamin B6 and other factors were associated with nodding syndrome. Our results argue against several previous causal hypotheses including Onchocerca volvulus. Instead, nodding syndrome may be caused by a complex interplay between multiple pathogens and nutrient levels. Further studies need to confirm these associations and determine the direction of effect.

2.
Virus Evol ; 8(2): veac087, 2022.
Article in English | MEDLINE | ID: mdl-36325032

ABSTRACT

Metagenomic techniques have facilitated the discovery of thousands of viruses, yet because samples are often highly biodiverse, fundamental data on the specific cellular hosts are usually missing. Numerous gastrointestinal viruses linked to human or animal diseases are affected by this, preventing research into their medical or veterinary importance. Here, we developed a computational workflow for the prediction of viral hosts from complex metagenomic datasets. We applied it to seven lineages of gastrointestinal cressdnaviruses using 1,124 metagenomic datasets, predicting hosts of four lineages. The Redondoviridae, strongly associated to human gum disease (periodontitis), were predicted to infect Entamoeba gingivalis, an oral pathogen itself involved in periodontitis. The Kirkoviridae, originally linked to fatal equine disease, were predicted to infect a variety of parabasalid protists, including Dientamoeba fragilis in humans. Two viral lineages observed in human diarrhoeal disease (CRESSV1 and CRESSV19, i.e. pecoviruses and hudisaviruses) were predicted to infect Blastocystis spp. and Endolimax nana respectively, protists responsible for millions of annual human infections. Our prediction approach is adaptable to any virus lineage and requires neither training datasets nor host genome assemblies. Two host predictions (for the Kirkoviridae and CRESSV1 lineages) could be independently confirmed as virus-host relationships using endogenous viral elements identified inside host genomes, while a further prediction (for the Redondoviridae) was strongly supported as a virus-host relationship using a case-control screening experiment of human oral plaques.

3.
PLoS One ; 17(10): e0276323, 2022.
Article in English | MEDLINE | ID: mdl-36264933

ABSTRACT

BACKGROUND: The increasing interest to perform and investigate the efficacy of fecal microbiota transplantation (FMT) has generated an urge for feasible donor screening. We report our experience with stool donor recruitment, screening, follow-up, and associated costs in the context of clinical FMT trials. METHODS: Potential stool donors, aged between 18-65 years, underwent a stepwise screening process starting with an extensive questionnaire followed by feces and blood investigations. When eligible, donors were rescreened for MDROs and SARS-CoV-2 every 60-days, and full rescreening every 4-6 months. The costs to find and retain a stool donor were calculated. RESULTS: From January 2018 to August 2021, 393 potential donors underwent prescreening, of which 202 (51.4%) did not proceed primarily due to loss to follow-up, medication use, or logistic reasons (e.g. COVID-19 measures). 191 potential donors filled in the questionnaire, of which 43 (22.5%) were excluded. The remaining 148 candidates underwent parasitology screening: 91 (61.5%) were excluded, mostly due to Dientamoeba fragilis and/or high amounts of Blastocystis spp. After additional feces investigations 18/57 (31.6%) potential donors were excluded (mainly for presence of Helicobacter Pylori and ESBL-producing organisms). One donor failed serum testing. Overall, 38 out of 393 (10%) potential donors were enrolled. The median participation time of active stool donors was 13 months. To recruit 38 stool donors, €64.112 was spent. CONCLUSION: Recruitment of stool donors for FMT is challenging. In our Dutch cohort, failed eligibility of potential donors was often caused by the presence of the protozoa Dientamoeba fragilis and Blastocystis spp.. The exclusion of potential donors that carry these protozoa, especially Blastocystis spp., is questionable and deserves reconsideration. High-quality donor screening is associated with substantial costs.


Subject(s)
COVID-19 , Clostridium Infections , Humans , Adolescent , Young Adult , Adult , Middle Aged , Aged , Fecal Microbiota Transplantation , Donor Selection , SARS-CoV-2 , Feces
4.
J Clin Virol ; 139: 104821, 2021 06.
Article in English | MEDLINE | ID: mdl-33882373

ABSTRACT

BACKGROUND: Detecting SARS-CoV-2 antibodies may help to diagnose COVID-19. Head-to-head validation of different types of immunoassays in well-characterized cohorts of hospitalized patients remains needed. METHODS: We validated three chemiluminescence immunoassays (CLIAs) (Liaison, Elecsys, and Abbott) and one single molecule array assay (SIMOA) (Quanterix) for automated analyzers, one rapid immunoassay RIA (AllTest), and one ELISA (Wantai) in parallel in first samples from 126 PCR confirmed COVID-19 hospitalized patients and 158 pre-COVID-19 patients. Specificity of the AllTest was also tested in 106 patients with confirmed parasitic and dengue virus infections. Specificity of the Wantai assay was not tested due to limitations in sample volumes. RESULTS: Overall sensitivity in first samples was 70.6 % for the Liaison, 71.4 % for the Elecsys, 75.4 % for the Abbott, 70.6 % for the Quanterix, 77.8 % for the AllTest, and 88.9 % for the Wantai assay, respectively. Sensitivity was between 77.4 % (Liaison) and 94.0 % (Wantai) after 10 dpso. No false positive results were observed for the Elecsys and Abbott assays. Specificity was 91.1 % for the Quanterix, 96.2 % for the Liaison, and 98.1 % for the AllTest assay, respectively. CONCLUSION: We conclude that low sensitivity of all immunoassays limits their use early after onset of illness in diagnosing COVID-19 in hospitalized patients. After 10 dpso, the Wantai ELISA has a relatively high sensitivity, followed by the point-of-care AllTest RIA that compares favorably with automated analyzer immunoassays.


Subject(s)
Antibodies, Viral/blood , COVID-19/diagnosis , Immunoassay/methods , SARS-CoV-2/immunology , Adult , Aged , Aged, 80 and over , COVID-19 Serological Testing , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Sensitivity and Specificity
5.
mSystems ; 6(2)2021 Mar 16.
Article in English | MEDLINE | ID: mdl-33727397

ABSTRACT

Bacterial microbiota play a critical role in mediating local and systemic immunity, and shifts in these microbial communities have been linked to impaired outcomes in critical illness. Emerging data indicate that other intestinal organisms, including bacteriophages, viruses of eukaryotes, fungi, and protozoa, are closely interlinked with the bacterial microbiota and their host, yet their collective role during antibiotic perturbation and critical illness remains to be elucidated. We employed multi-omics factor analysis (MOFA) to systematically integrate the bacterial (16S rRNA), fungal (intergenic transcribed spacer 1 rRNA), and viral (virus discovery next-generation sequencing) components of the intestinal microbiota of 33 critically ill patients with and without sepsis and 13 healthy volunteers. In addition, we quantified the absolute abundances of bacteria and fungi using 16S and 18S rRNA PCRs and characterized the short-chain fatty acids (SCFAs) butyrate, acetate, and propionate using nuclear magnetic resonance spectroscopy. We observe that a loss of the anaerobic intestinal environment is directly correlated with an overgrowth of aerobic pathobionts and their corresponding bacteriophages as well as an absolute enrichment of opportunistic yeasts capable of causing invasive disease. We also observed a strong depletion of SCFAs in both disease states, which was associated with an increased absolute abundance of fungi with respect to bacteria. Therefore, these findings illustrate the complexity of transkingdom changes following disruption of the intestinal bacterial microbiome.IMPORTANCE While numerous studies have characterized antibiotic-induced disruptions of the bacterial microbiome, few studies describe how these disruptions impact the composition of other kingdoms such as viruses, fungi, and protozoa. To address this knowledge gap, we employed MOFA to systematically integrate viral, fungal, and bacterial sequence data from critically ill patients (with and without sepsis) and healthy volunteers, both prior to and following exposure to broad-spectrum antibiotics. In doing so, we show that modulation of the bacterial component of the microbiome has implications extending beyond this kingdom alone, enabling the overgrowth of potentially invasive fungi and viruses. While numerous preclinical studies have described similar findings in vitro, we confirm these observations in humans using an integrative analytic approach. These findings underscore the potential value of multi-omics data integration tools in interrogating how different components of the microbiota contribute to disease states. In addition, our findings suggest that there is value in further studying potential adjunctive therapies using anaerobic bacteria or SCFAs to reduce fungal expansion after antibiotic exposure, which could ultimately lead to improved outcomes in the intensive care unit (ICU).

6.
Pediatr Infect Dis J ; 40(4): 279-283, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33181781

ABSTRACT

BACKGROUND: Dientamoeba fragilis in children has been associated with gastrointestinal symptoms, like abdominal pain and diarrhea. The mechanism underlying these symptoms in children with D. fragilis remains unclear. We hypothesized that concomitant microbial alterations, which have been described in other parasitic infections, may be associated with gastrointestinal symptoms in D. fragilis. METHODS: In this case-control study performed in 2 centers, 19 children referred to a pediatrician because of gastrointestinal symptoms and with a positive fecal PCR for D. fragilis were included as cases. We included 19 healthy children as controls and matched for age and gender, selected from an existing cohort of 63 children. A PCR for D. fragilis was performed on fecal samples of the 19 controls to assess D. fragilis carriership in this asymptomatic group. Microbiota was analyzed with the IS-pro technique, and the intestinal microbiota composition and diversity were compared between the 2 groups. RESULTS: Microbiota of children with D. fragilis and gastrointestinal symptoms did not significantly differ in terms of composition and diversity compared with controls, both on phylum and species level. In the asymptomatic controls, a positive fecal PCR for D. fragilis was found in 16 of 19 (84.2%). CONCLUSION: Intestinal microbiota does not seem to play a key role in the presence of clinical symptoms in children with D. fragilis. The pathogenicity of D. fragilis and pathophysiologic pathways underlying the development of gastrointestinal symptoms remains yet to be clarified.


Subject(s)
Dientamoeba/genetics , Dientamoebiasis/parasitology , Gastrointestinal Diseases/parasitology , Gastrointestinal Microbiome/genetics , Abdominal Pain , Case-Control Studies , Child , Child, Preschool , Cohort Studies , Diarrhea/parasitology , Dientamoeba/pathogenicity , Feces/parasitology , Genetic Variation , Humans
7.
Nat Commun ; 11(1): 4620, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32934242

ABSTRACT

Metagenomic techniques have enabled genome sequencing of unknown viruses without isolation in cell culture, but information on the virus host is often lacking, preventing viral characterisation. High-throughput methods capable of identifying virus hosts based on genomic data alone would aid evaluation of their medical or biological relevance. Here, we address this by linking metagenomic discovery of three virus families in human stool samples with determination of probable hosts. Recombination between viruses provides evidence of a shared host, in which genetic exchange occurs. We utilise networks of viral recombination to delimit virus-host clusters, which are then anchored to specific hosts using (1) statistical association to a host organism in clinical samples, (2) endogenous viral elements in host genomes, and (3) evidence of host small RNA responses to these elements. This analysis suggests two CRESS virus families (Naryaviridae and Nenyaviridae) infect Entamoeba parasites, while a third (Vilyaviridae) infects Giardia duodenalis. The trio supplements five CRESS virus families already known to infect eukaryotes, extending the CRESS virus host range to protozoa. Phylogenetic analysis implies CRESS viruses infecting multicellular life have evolved independently on at least three occasions.


Subject(s)
Entamoeba/virology , Giardia/virology , Adult , Cohort Studies , Feces/parasitology , Feces/virology , Female , Genome, Viral , Host Specificity , Humans , Male , Middle Aged , Phylogeny , Virus Physiological Phenomena , Viruses/classification , Viruses/genetics , Young Adult
8.
Expert Rev Gastroenterol Hepatol ; 14(4): 231-242, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32155096

ABSTRACT

Introduction: The presence of D. fragilis in feces is characterized by an asymptomatic carrier ship to a spectrum of gastrointestinal symptoms. However, a causal relationship remains to be elucidated. In this systematic review, we aimed to evaluate the relationship between the eradication of D. fragilis and symptoms to establish the strength of evidence that D. fragilis in symptomatic children warrants antibiotic treatment.Areas covered: This systematic review covers a challenge in daily clinical practice. Is it necessary to test for D. fragilis in children with gastrointestinal symptoms and does a positive fecal PCR test warrant treatment?Expert opinion: Testing for D. fragilis seems justified in a selection of children with persistent unexplained chronic abdominal pain and diarrhea. Treatment of D. fragilis should be withhold until other causes like celiac disease have been excluded. Both microscopic and Real Time-PCR methods (or a combination of the two) can be used for diagnosis. Paromomycin or clioquinol are antibiotics of choice based on their small spectrum of activity, fewer side effects, and better eradication rates than metronidazole. Future randomized studies, with strict inclusion criteria, appropriate diagnostic testing, and doses of antibiotics based on bodyweight are warranted.


Subject(s)
Dientamoebiasis/diagnosis , Dientamoebiasis/drug therapy , Abdominal Pain/drug therapy , Abdominal Pain/etiology , Abdominal Pain/parasitology , Child , Diagnosis, Differential , Diarrhea/drug therapy , Diarrhea/etiology , Diarrhea/parasitology , Dientamoeba/isolation & purification , Dientamoebiasis/complications , Dientamoebiasis/parasitology , Feces/parasitology , Humans , Real-Time Polymerase Chain Reaction , Treatment Outcome
9.
Clin Infect Dis ; 71(10): 2630-2636, 2020 12 17.
Article in English | MEDLINE | ID: mdl-31728525

ABSTRACT

BACKGROUND: Patients with multiple recurrent Clostridioides difficile infections (rCDI) are treated with fecal microbiota transplantation (FMT), using feces provided by healthy donors. Blastocystis colonization of donors is considered an exclusion criterion, whereas its pathogenicity is still under debate. METHODS: The introduction of molecular screening for Blastocystis sp. at our stool bank identified 2 donors with prior negative microscopies but positive polymerase chain reactions (PCRs). Potential transmission of Blastocystis sp. to patients was assessed on 16 fecal patient samples, pre- and post-FMT, by PCR and subtype (ST) analyses. In addition, clinical outcomes for the treatment of rCDI (n = 31), as well as the development of gastrointestinal symptoms, were assessed. RESULTS: There was 1 donor who carried Blastocystis ST1, and the other contained ST3. All patients tested negative for Blastocystis prior to FMT. With a median diagnosis at 20.5 days after FMT, 8 of 16 (50%) patients developed intestinal colonization with Blastocystis, with identical ST sequences as their respective donors. Blastocystis-containing fecal suspensions were used to treat 31 rCDI patients, with an FMT success rate of 84%. This success rate was not statistically different from patients transferred with Blastocystis sp.-negative donor feces (93%, 76/82). Patients transferred with Blastocystis sp.-positive donor feces did not report any significant differences in bowel complaints in the first week, after 3 weeks, or in the months following FMT. CONCLUSIONS: We demonstrated the first transmission of Blastocystis ST1 and ST3 from donors to patients by FMT. This did not result in gastrointestinal symptomatology or have any significant effect on rCDI treatment outcomes.


Subject(s)
Blastocystis , Clostridioides difficile , Clostridium Infections , Blastocystis/genetics , Fecal Microbiota Transplantation , Feces , Humans , Treatment Outcome
10.
Ned Tijdschr Geneeskd ; 1632019 08 07.
Article in Dutch | MEDLINE | ID: mdl-31424703

ABSTRACT

In the Netherlands, approximately 200 to 300 patients are diagnosed with imported malaria every year. The symptoms of malaria are non-specific. The current gold standard for malaria diagnostics is to conduct a thick and thin blood smear. New diagnostic techniques are increasingly applied. At present, the treatment of uncomplicated malaria consists of an artemisinin-based combination therapy (ACT). An alternative treatment for malaria caused by P. vivax,P. knowlesi,P. ovale and P. malariae in the Netherlands is chloroquine. Severe malaria is treated with artesunate intravenously, followed by a full three-day course of oral ACT. Uncomplicated malaria during pregnancy is treated with an ACT (e.g. artemether-lumefantrine) and severe malaria with artesunate intravenously, the latter followed by a full three-day course of oral ACT. There is currently no malaria vaccine available for travellers.


Subject(s)
Antimalarials/therapeutic use , Malaria/ethnology , Pregnancy Complications, Parasitic/epidemiology , Travel , Female , Humans , Malaria/drug therapy , Malaria Vaccines , Netherlands/epidemiology , Pregnancy , Pregnancy Complications, Parasitic/drug therapy
11.
Ned Tijdschr Geneeskd ; 1632019 05 24.
Article in Dutch | MEDLINE | ID: mdl-31166095

ABSTRACT

In the Netherlands, approximately 200 to 300 patients are diagnosed with imported malaria every year. The symptoms of malaria are non-specific. The current gold standard for malaria diagnostics is to conduct a thick and thin blood smear. New diagnostic techniques are increasingly applied. At present, the treatment of uncomplicated malaria consists of an artemisinin-based combination therapy (ACT). An alternative treatment for malaria caused by P. vivax,P. knowlesi,P. ovale and P. malariae in the Netherlands is chloroquine. Severe malaria is treated with artesunate intravenously, followed by a full three-day course of oral ACT. Uncomplicated malaria during pregnancy is treated with an ACT (e.g. artemether-lumefantrine) and severe malaria with artesunate intravenously, the latter followed by a full three-day course of oral ACT. There is currently no malaria vaccine available for travellers.


Subject(s)
Antimalarials/therapeutic use , Artemisinins/therapeutic use , Malaria/drug therapy , Travel , Artemether/therapeutic use , Artemether, Lumefantrine Drug Combination , Chloroquine/therapeutic use , Drug Combinations , Drug Therapy, Combination , Female , Humans , Malaria/diagnosis , Malaria Vaccines/therapeutic use , Male , Netherlands , Pregnancy
12.
Psychol Med ; 49(10): 1608-1623, 2019 07.
Article in English | MEDLINE | ID: mdl-31010440

ABSTRACT

Unnatural causes of death due to traffic accidents (TA) and suicide attempts (SA) constitute a major burden on global health, which remained stable in the last decade despite widespread efforts of prevention. Recently, latent infection with Toxoplasma gondii (T. gondii) has been suggested to be a biological risk factor for both TA and SA. Therefore, a systematic search concerning the relationship of T. gondii infection with TA and/or SA according to PRISMA guidelines in Medline, Pubmed and PsychInfo was conducted collecting papers up to 11 February 2019 (PROSPERO #CRD42018090206). The random-effect model was applied and sensitivity analyses were subsequently performed. Lastly, the population attributable fraction (PAF) was calculated. We found a significant association for antibodies against T. gondii with TA [odds ratio (OR) = 1.69; 95% confidence interval (CI) 1.20-2.38, p = 0.003] and SA (OR = 1.39; 95% CI 1.10-1.76, p = 0006). Indication of publication bias was found for TA, but statistical adjustment for this bias did not change the OR. Heterogeneity between studies on SA was partly explained by type of control population used (ORhealthy controls = 1.9, p < 0.001 v. ORpsychiatric controls = 1.06, p = 0.87) and whether subjects with schizophrenia only were analysed (ORschizophrenia = 0.87, p = 0.62 v. ORvarious = 1.8, p < 0.001). The association was significantly stronger with higher antibody titres in TA and in studies that did not focus on schizophrenia subjects concerning SA. PAF of a T. gondii infection was 17% for TA and 10% for SA. This indicates that preventing T. gondii infection may play a role in the prevention of TA or SA, although uncertainty remains whether infection and outcome are truly causally related.


Subject(s)
Accidents, Traffic/statistics & numerical data , Suicide, Attempted/statistics & numerical data , Toxoplasmosis/epidemiology , Humans
13.
Travel Med Infect Dis ; 27: 87-91, 2019.
Article in English | MEDLINE | ID: mdl-29929001

ABSTRACT

INTRODUCTION: A lack of prospective and longitudinal data on pre- and post-travel carriage of Blastocystis spp. complicates interpretation of a positive test post-travel. Therefore we studied dynamics of Blastocystis carriage in a cohort of Dutch travellers. METHODS: From the prospective, multicentre COMBAT study among 2001 Dutch travellers, a subset of 491 travellers was selected based on travel destination to 7 subregions (70 or 71 travellers each). Faecal samples taken directly before and after travel were screened for Blastocystis with qPCR, followed, when positive, by sequence analysis to determine subtypes. RESULTS: After exclusion of 12 samples with missing samples or inhibited qPCR-reactions, stool samples of 479 travellers were analysed. Before travel, 174 of them (36.3%) carried Blastocystis and in most of these, the same subtype was persistently carried. However, in 48/174 of those travellers (27.6%; CI95 20.8-36.6%) no Blastocystis or a different subtype was detected in the post-travel sample, indicating loss of Blastocystis during travel. Only 26 (5.4%; CI95 3.7%-8.0%) of all travellers acquired Blastocystis, including two individuals that were already positive for Blastocystis before travel but acquired a different subtype during travel. DISCUSSION: This study shows that Blastocystis carriage in travellers is highly dynamic. The observed acquisition and loss of Blastocystis could either be travel-related or reflect the natural course of Blastocystis carriage. We demonstrate that the majority of Blastocystis detected in post-travel samples were already carried before travel.


Subject(s)
Blastocystis Infections/epidemiology , Carrier State/parasitology , Feces/parasitology , Travel , Adult , Aged , Aged, 80 and over , Blastocystis/genetics , Carrier State/epidemiology , Female , Healthy Volunteers , Humans , Longitudinal Studies , Male , Middle Aged , Netherlands/epidemiology , Prospective Studies , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Young Adult
14.
PLoS One ; 13(5): e0197770, 2018.
Article in English | MEDLINE | ID: mdl-29847574

ABSTRACT

BACKGROUND: Despite the considerable burden of helminth infections in developing countries and increasing international travel, little is known about the risks of infection for travelers. OBJECTIVE: We studied the attack and incidence rate of serology confirmed strongyloidiasis, filariasis, and toxocariasis among long-term travelers and associated factors. A second objective was to evaluate eosinophilia as a positive/negative predictive value (PPV/NPV) for a recent helminth infection. METHODS: From 2008 to 2011, clients of the Public Health Service travel clinic planning travel to (sub)tropical countries for 12-52 weeks were invited to participate in a prospective study. Participants kept a weekly diary, recording itinerary, symptoms, and physician visits during travel and completed a post-travel questionnaire. Pre- and post-travel blood samples were serologically tested for the presence of IgG antibodies against Schistosoma species, Strongyloides stercoralis, filarial species, and Toxacara species and were used for a blood cell count. Factors associated with recent infection were analyzed using Poisson regression. Differences among groups of travelers were studied using chi square tests. RESULTS: For the 604 participants, median age was 25 years (interquartile range [IQR]: 23-29), 36% were male, median travel duration was 20 weeks (IQR: 15-25), and travel purpose was predominantly tourism (62%). Destinations were Asia (45%), Africa (18%), and the Americas (37%). Evidence of previous infection was found in 13/604 participants: antibodies against Schistosoma spp. in 5 (0.8%), against S.stercoralis in 3 (0.5%), against filarial species in 4 (0.7%), and against Toxocara spp. in 1 (0.2%). Ten recent infections were found in 9 participants (3, 1, 6, 0 cases, in the above order), making the attack rates 0.61, 0.17, 1.1 and 0, and the incidence rates per 1000 person-months 1.5, 0.34, 2.6 and 0. The overall PPV and NPV of eosinophila for recent infection were 0 and 98%, respectively. CONCLUSIONS: The risk of the helminth infections under study in this cohort of long-term travelers was low. Routine screening for eosinophilia appeared not to be of diagnostic value.


Subject(s)
Filariasis/epidemiology , Schistosomiasis/epidemiology , Strongyloidiasis/epidemiology , Toxocariasis/epidemiology , Adult , Antibodies, Helminth/metabolism , Eosinophilia/diagnosis , Female , Filariasis/immunology , Humans , Immunoglobulin G/metabolism , Incidence , Male , Netherlands/epidemiology , Poisson Distribution , Prospective Studies , Schistosomiasis/immunology , Strongyloidiasis/immunology , Toxocariasis/immunology , Travel-Related Illness , Young Adult
15.
Am J Trop Med Hyg ; 99(1): 90-93, 2018 07.
Article in English | MEDLINE | ID: mdl-29692301

ABSTRACT

Microsporidia are protists close to the kingdom of fungi that may cause eye infections. Most cases are reported in Asia and affect both immunocompromised and immunocompetent patients. Here, we report a rare case of microsporidial keratoconjunctivitis in an immunocompetent French patient 3 weeks after returning from India. In our patient, Weber trichrome staining of conjunctival scrapings revealed rounded elements approximately 1-3 µm in size. Conventional polymerase chain reaction analysis by ribosomal RNA subunit sequencing showed 100% identity with Vittaforma corneae. Treatment by corneal debridement combined with fluoroquinolone eye drops allowed complete resolution of the lesions. Although rare, ocular microsporidiosis should be investigated in a patient who is native to Asia or has returned from an endemic area and presents with keratoconjunctivitis of undetermined etiology.


Subject(s)
Antifungal Agents/therapeutic use , Eye Infections, Fungal/diagnosis , Fluoroquinolones/therapeutic use , Keratoconjunctivitis/diagnosis , Microsporidiosis/diagnosis , Cornea/drug effects , Cornea/microbiology , Cornea/pathology , Cornea/surgery , Debridement/methods , Eye Infections, Fungal/drug therapy , Eye Infections, Fungal/microbiology , Eye Infections, Fungal/surgery , France , Humans , India , Keratoconjunctivitis/drug therapy , Keratoconjunctivitis/microbiology , Keratoconjunctivitis/surgery , Male , Microsporidiosis/drug therapy , Microsporidiosis/microbiology , Microsporidiosis/surgery , Middle Aged , Travel , Vittaforma/drug effects , Vittaforma/growth & development , Vittaforma/pathogenicity
16.
Travel Med Infect Dis ; 19: 33-36, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28842214

ABSTRACT

BACKGROUND: Limited prospective data are available on the acquisition of viral, bacterial and parasitic diarrhoeagenic agents by healthy individuals during travel. METHODS: To determine the frequency of travel associated acquisition of 19 pathogens in 98 intercontinental travellers, qPCR was used to detect 8 viral pathogens, 6 bacterial enteric pathogens and 5 parasite species in faecal samples collected immediately before and after travel. RESULTS: We found high pre-travel carriage rates of Blastocystis spp. and Dientamoeba fragilis of 32% and 19% respectively. Pre-travel prevalences of all other tested pathogens were below 3%. Blastocystis spp. (10%), Plesiomonas shigelloides (7%), D. fragilis (6%) and Shigella spp. (5%) were the most frequently acquired pathogens and acquisition of enteral viruses and hepatitis E virus in this relatively small group of travellers was rare or non-existent. CONCLUSIONS: Our findings suggest that the role of viruses as the cause of persisting traveller's diarrhoea is limited and bacterial pathogens are more likely as a cause of traveller's diarrhoea. The substantial proportion of travellers carrying Blastocystis spp. and D. fragilis before travel warrants cautious interpretation of positive samples in returning travellers with gastrointestinal complaints.


Subject(s)
Diarrhea , Travel-Related Illness , Cohort Studies , Diarrhea/microbiology , Diarrhea/parasitology , Diarrhea/virology , Enterobacteriaceae Infections/epidemiology , Enterovirus Infections/epidemiology , Feces/microbiology , Feces/parasitology , Feces/virology , Humans , Netherlands/epidemiology , Parasitic Diseases/epidemiology , Prevalence , Prospective Studies
17.
Acta Trop ; 173: 90-96, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28587839

ABSTRACT

In Turkey, the main causative agents are Leishmania tropica (L. tropica) and Leishmania infantum (L. infantum) for cutaneous leishmaniasis (CL) and L. infantum for visceral leishmaniasis (VL). In this study, we investigated leishmaniasis cases caused by L. donovani and established animal models for understanding its tropism in in vivo conditions. Clinical samples (lesion aspirations and bone marrow) obtained from CL/VL patients were investigated using parasitological (smear/NNN) and DNA-based techniques. For species identification, a real time ITS1-PCR was performed using isolates and results were confirmed by hsp70 PCR-N/sequencing and cpb gene PCR/sequencing in order to reveal Leishmania donovani and Leishmania infantum discrimination. Clinical materials from CL and VL patients were also inoculated into two experimental groups (Group CL and Group VL) of Balb/C mice intraperitoneally for creating clinical picture of Turkish L. donovani strains. After 45days, the samples from visible sores of the skin were taken, and spleens and livers were removed. Measurements of the internal organs were done and touch preparations were prepared for checking the presence of amastigotes. The strains were isolated from all patients and amastigotes were seen in all smears of the patients, and then isolates were immediately stored in liquid nitrogen. In real time ITS1-PCR, the melting temperatures of all samples were out of range of L. infantum, L. tropica and L. major. Sequencing of hsp70 PCR-N showed that all isolates highly identical to previously submitted L. donovani sequences in GenBank, and cpb gene sequencing showed five isolates had longer cpbF allele, whereas one isolate contained a mixed sequence of both cpbF and cpbE. All mice in both experimental groups became infected. Compared to controls, the length and width of both liver and spleen were significantly elevated (p<0.001) in both groups of mice. However, the weight of the liver increased significantly in all mice whereas the weight of spleen increased only in VL group. Amastigotes were also seen in all touch preparations prepared from skin sores, spleen and liver. L. donovani strain was isolated from autocutaneous a VL patient first time in Turkey. Animal models using clinical samples were successfully established and important clinical differences of the isolated strains were observed.


Subject(s)
Leishmania donovani/isolation & purification , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Visceral/parasitology , Animals , Humans , Leishmania donovani/genetics , Leishmaniasis, Cutaneous/epidemiology , Leishmaniasis, Visceral/epidemiology , Male , Mice , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction , Skin/parasitology , Skin/pathology , Turkey/epidemiology
19.
Genom Data ; 4: 22-3, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26484170

ABSTRACT

The intestinal protistan parasite Blastocystis is characterized by an extensive genetic variability with 17 subtypes (ST1-ST17) described to date. Only the whole genome of a human ST7 isolate was previously sequenced. Here we report the draft genome sequence of Blastocystis ST4-WR1 isolated from a laboratory rodent at Singapore.

20.
Ned Tijdschr Geneeskd ; 159: A8774, 2015.
Article in Dutch | MEDLINE | ID: mdl-25990333

ABSTRACT

Malaria usually begins with nonspecific symptoms which could easily be ascribed to other common febrile illnesses and hence be missed. If left untreated, it can lead to severe complicated malaria and death. We describe a case of a 43-year-old male patient with fever who recently returned from West Africa. Initially he was treated for pneumonia; however, after 4 days his symptoms deteriorated and he was diagnosed with severe complicated falciparum malaria for which he was admitted to the ICU. We advocate prompt exclusion of malaria in every patient presenting with fever in the three months following return from an endemic region, and remaining vigilant for an extended period.


Subject(s)
Fever/etiology , Malaria, Falciparum/diagnosis , Travel , Adult , Africa, Western , Fever/diagnosis , Humans , Malaria, Falciparum/epidemiology , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...