1
|
Ogundare ST, Fasina FO, Makumbi JP, van der Zel GA, Geertsma PF, Kock MM, Smith AM, Ehlers MM. Epidemiology and antimicrobial resistance profiles of pathogenic Escherichia coli from commercial swine and poultry abattoirs and farms in South Africa: A One Health approach. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175705. [PMID: 39181266 DOI: 10.1016/j.scitotenv.2024.175705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Pathogenic Escherichia coli (PEC) are important foodborne bacteria that can cause severe illness in humans. The PECs thrive within the intestines of humans as well as animals and may contaminate multiple ecosystems, including food and water, via faecal transmission. Abattoir and farm employees are at high risk of PEC exposure, which could translate to community risk through person-to-person contact. To determine the epidemiology and resistome of PECs in Gauteng and Limpopo provinces of South Africa, 198 swine faecal samples, 220 poultry cloacal swabs, 108 human hand swabs, 11 run-off water samples from abattoirs and farms were collected from four swine and five poultry commercial abattoirs and two swine farms. One effluent sample each was collected from four wastewater treatment plants (WWTP) and a tertiary hospital setting. Phenotypic and genotypic techniques were used including polymerase chain reaction, pulsed-field gel electrophoresis (PFGE) and whole genome sequencing (WGS). Results showed EHEC and EPEC prevalence was 4.1 % (22/542) and 20.8 % (113/542), respectively, with the O26 serogroup detected the most in PEC isolates. According to the PFGE dendrogram, isolates from poultry, human hand swabs and run-off water clustered together. Diverse virulence factors such as the novel stx2k subtype and eae genes were detected among the 36 representative PEC isolates according to WGS. The results showed that 66.7 % (24/36) of sequenced PECs presented with multi-drug resistance (MDR) to β-lactamase 13.9 % (5/36), aminoglycoside 61.1 % (22/36), tetracycline 41.7 % (15/36) and quinolones 38.9 % (14/36). No colistin nor carbapenem resistance was detected. Sequence types (STs) associated with MDR in this study were: ST752, ST189, ST206, ST10, ST48 and ST38. The findings highlight the threat of zoonotic pathogens to close human contacts and the need for enhanced surveillance to mitigate the spread of MDR foodborne PECs.
Collapse
Affiliation(s)
- Samuel T Ogundare
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| | - Folorunso O Fasina
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa; Food and Agriculture Organisation of the United Nations, FAO Headquarters, Rome, Italy
| | - John-Paul Makumbi
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Gerbrand A van der Zel
- Gauteng Department of Agriculture, Rural Development and Environment, Pretoria, South Africa
| | - Peter F Geertsma
- Gauteng Department of Agriculture, Rural Development and Environment, Pretoria, South Africa
| | - Marleen M Kock
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; Department of Medical Microbiology, National Health Laboratory Service, Tshwane Academic Division, Pretoria, South Africa
| | - Anthony M Smith
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; Centre for Enteric Diseases, National Institute for Communicable Diseases, Division of the National Health Laboratory Service, Johannesburg, South Africa
| | - Marthie M Ehlers
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; Department of Medical Microbiology, National Health Laboratory Service, Tshwane Academic Division, Pretoria, South Africa
| |
Collapse
|
2
|
Schneeweiss Garber N, Bourgade Su PA, Lozano Guerrero G, Hernandez Salazar A, Aboitiz CM. Enteroaggregative and Enteroinvasive Escherichia coli as a Cause of Pediatric Acute Abdomen: A Report of Two Cases. Cureus 2024; 16:e68340. [PMID: 39355069 PMCID: PMC11444806 DOI: 10.7759/cureus.68340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2024] [Indexed: 10/03/2024] Open
Abstract
Abdominal pain stands as one of the foremost reasons for consultation among pediatric patients, presenting a diagnostic challenge owing to its diverse underlying causes. The manifestation of abdominal pain varies according to age, associated symptoms, and pain localization. While frequently self-limited, certain conditions exist that endanger life and require urgent intervention. Acute abdomen denotes severe, non-traumatic abdominal pain resulting from inflammatory, ischemic, obstructive, infectious, gynecological, or metabolic etiologies, warranting immediate therapeutic intervention. Infectious processes that mimic acute abdominal conditions are relatively uncommon. Consequently, the identification of infectious gastroenteritis as a probable etiology of acute abdomen can prevent unnecessary surgical interventions in patients. This report details two cases: a 14-year-old pediatric patient presenting with acute abdominal pain, in whom appendiceal involvement was ruled out through ultrasonographic and computed tomography (CT) examinations, confirming the presence of enteroaggregative Escherichia coli, and a 10-year-old pediatric patient presenting with a sudden onset of abdominal pain. CT findings revealed an appendiceal fecalith without concurrent inflammation but accompanied by mesenteric adenitis. Even though conservative treatment did not improve the pain, it was later determined that the patient was a carrier of enteroinvasive E. coli. In both cases, antimicrobial treatment with rifaximin 200 mg every eight hours was administered, leading to the resolution of the conditions without the need for hospital readmission or additional therapy. Infectious conditions stemming from enteroaggregative and enteroinvasive E. coli can mimic acute abdomen and should be regarded as potential infectious etiologies when other more common causes have been ruled out.
Collapse
Affiliation(s)
| | - Paul A Bourgade Su
- Faculty of Health Sciences, Universidad Anáhuac México, Mexico City, MEX
| | | | | | - Carlos Manuel Aboitiz
- Pediatric Cardiology, Hospital Angeles, Mexico City, MEX
- Cardiology, Instituto Nacional de Enfermedades Respiratorias, Mexico City, MEX
| |
Collapse
|
3
|
Van Nederveen V, Melton-Celsa A. Extracellular components in enteroaggregative Escherichia coli biofilm and impact of treatment with proteinase K, DNase or sodium metaperiodate. Front Cell Infect Microbiol 2024; 14:1379206. [PMID: 38938878 PMCID: PMC11209426 DOI: 10.3389/fcimb.2024.1379206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/23/2024] [Indexed: 06/29/2024] Open
Abstract
Enteroaggregative E. coli (EAEC) is a major cause of diarrhea worldwide. EAEC are highly adherent to cultured epithelial cells and make biofilms. Both adherence and biofilm formation rely on the presence of aggregative adherence fimbriae (AAF). We compared biofilm formation from two EAEC strains of each of the five AAF types. We found that AAF type did not correlate with the level of biofilm produced. Because the composition of the EAEC biofilm has not been fully described, we stained EAEC biofilms to determine if they contained protein, carbohydrate glycoproteins, and/or eDNA and found that EAEC biofilms contained all three extracellular components. Next, we assessed the changes to the growing or mature EAEC biofilm mediated by treatment with proteinase K, DNase, or a carbohydrate cleavage agent to target the different components of the matrix. Growing biofilms treated with proteinase K had decreased biofilm staining for more than half of the strains tested. In contrast, although sodium metaperiodate only altered the biofilm in a quantitative way for two strains, images of biofilms treated with sodium metaperiodate showed that the EAEC were more spread out. Overall, we found variability in the response of the EAEC strains to the treatments, with no one treatment producing a biofilm change for all strains. Finally, once formed, mature EAEC biofilms were more resistant to treatment than biofilms grown in the presence of those same treatments.
Collapse
Affiliation(s)
- Viktoria Van Nederveen
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Angela Melton-Celsa
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
4
|
Haugan I, Husby MG, Skjøtskift B, Aamnes Mostue D, Brun A, Olsen LC, Simpson MR, Lange H, Afset JE. Enteroaggregative Escherichia coli in mid-Norway: A prospective, case control study. PLoS One 2024; 19:e0301625. [PMID: 38635516 PMCID: PMC11025732 DOI: 10.1371/journal.pone.0301625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 03/19/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND The use of molecular methods has led to increased detection of Enteroaggregative Escherichia coli (EAEC) in faecal samples. Studies have yielded conflicting results regarding the clinical relevance of this finding. The objective of this study was to investigate the prevalence of EAEC in faecal samples from patients with diarrhoea and healthy controls and describe characteristics of EAEC positive persons. METHODS From March 1st, 2017 to February 28th, 2019, we investigated all consecutive faecal samples from patients with diarrhoea received at the laboratory and collected faecal samples from randomly invited healthy controls from mid-Norway. Real-time multiplex PCR was used for detection of bacterial, viral, and parasitic pathogens. We registered sex, age, urban versus non-urban residency, and travel history for all participants. Statistical analyses were performed with Pearson chi-squared test, Kruskal-Wallis test, and Mann-Whitney U test. RESULTS We identified EAEC in 440 of 9487 (4.6%) patients with diarrhoea and 8 of 375 (2.2%) healthy controls. The EAEC prevalence was 19.1% among those with diarrhoea and recent foreign travel and 2.2% in those without travel history independent of diarrhoea. Concomitant pathogens were detected in 64.3% of EAEC-positive patients with diarrhoea. The median age was 28.5 in those with EAEC-positive diarrhoea and 38 in those with EAEC-negative diarrhoea (p <0.01). In patients with diarrhoea, travel was reported in 72% of those with EAEC and concomitant pathogens, and 54% and 12% in those with only EAEC and no EAEC, respectively (p <0.01). CONCLUSIONS EAEC was a common detection, particularly in patients with diarrhoea and recent international travel, and was found together with other intestinal pathogens in the majority of cases. Our results suggest that domestically acquired EAEC is not associated with diarrhoea. Patients with EAEC-positive diarrhoea and concomitant pathogens were young and often reported recent travel history compared to other patients with diarrhoea.
Collapse
Affiliation(s)
- Ingvild Haugan
- Department of Medical Microbiology, Clinic of Laboratory Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marit Gudrun Husby
- Department of Medical Microbiology, Clinic of Laboratory Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Bjørg Skjøtskift
- Department of Medical Microbiology, Clinic of Laboratory Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Dorothea Aamnes Mostue
- Department of Medical Microbiology, Clinic of Laboratory Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Andreas Brun
- Department of Medical Microbiology, Clinic of Laboratory Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Lene Christin Olsen
- Department of Medical Microbiology, Clinic of Laboratory Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Clinical Research Unit Central Norway, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Heidi Lange
- Norwegian Institute of Public Health, Oslo, Norway
| | - Jan Egil Afset
- Department of Medical Microbiology, Clinic of Laboratory Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
5
|
Akinlabi OC, Nwoko ESQ, Dada RA, Ekpo S, Omotuyi A, Nwimo CC, Adepoju A, Popoola O, Dougan G, Thomson NR, Okeke IN. Epidemiology and Risk Factors for Diarrheagenic Escherichia coli Carriage among Children in Northern Ibadan, Nigeria. Am J Trop Med Hyg 2023; 109:1223-1232. [PMID: 37903436 PMCID: PMC10793065 DOI: 10.4269/ajtmh.22-0618] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 08/14/2023] [Indexed: 11/01/2023] Open
Abstract
Diarrhea is a leading cause of childhood morbidity in Africa, but few studies, focus on bacterial diarrheal etiology including multicountry studies that typically excluded Nigeria. We collected stool specimens from 477 children under 5 years of age, 120 with diarrhea, who were enrolled in our prospective case-control study between November 2015 and August 2019. All were attending primary health clinics on the northern outskirts of Ibadan. Up to 10 Escherichia coli isolates were obtained per specimen, and at least three of them were sequenced using Illumina whole-genome sequence technology. Genomes were assembled using SPAdes and evaluated for quality using QUAST. VirulenceFinder was used to identify virulence genes. The microbiological quality of water from 14 wells within the study area was assessed using total and coliform counts. Diarrheagenic E. coli (DEC) were isolated from 79 (65.8%) cases and 217 (60.8%) control children. A number of hybrid DEC pathotypes, Salmonella spp., Yersinia spp., and all DEC pathotypes except Shiga toxin-producing E. coli were detected, but no pathogen showed association with disease (P > 0.05). Enterotoxigenic E. coli were more commonly recovered from children without diarrhea aged below 6 months but exclusively detected in children with diarrhea aged over 9 months. Temporally linked, genetically similar enteroaggregative E. coli were isolated from children in different households in eight instances. No well water sample drawn in the study was potable. Children in northern Ibadan were commonly colonized with DEC. Access to water, proper sanitation, and vaccination against the prevailing pathogens may be critical for protecting children from the less overt consequences of enteric pathogen carriage.
Collapse
Affiliation(s)
- Olabisi C. Akinlabi
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Oyo, Nigeria
| | - El-shama Q. Nwoko
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Oyo, Nigeria
| | - Rotimi A. Dada
- Medical Laboratory Science Program, College of Health Sciences, Bowen University, Iwo, Nigeria
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Ahmadu Bello University, Zaria, Nigeria
| | - Stella Ekpo
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Oyo, Nigeria
| | - Adeola Omotuyi
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Oyo, Nigeria
| | - Chukwuemeka C. Nwimo
- Department of Clinical Medicine, College of Medicine, University of Ibadan, Oyo, Nigeria
| | - Akinlolu Adepoju
- Department of Pediatrics, College of Medicine, University of Ibadan, Oyo, Nigeria
| | - Oluwafemi Popoola
- Department of Community Medicine, Faculty of Public Health, College of Medicine, University of Ibadan, Oyo, Nigeria
| | - Gordon Dougan
- Wellcome Sanger Institute, Saffron Walden, United Kingdom
- Department of Medicine, University of Cambridge, United Kingdom
| | | | - Iruka N. Okeke
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Oyo, Nigeria
| |
Collapse
|
6
|
Izquierdo-Vega JA, Castillo-Juarez RJ, Sánchez-Gutiérrez M, Ares MA, De La Cruz MA. A Mini-Review of Enteroaggregative Escherichia coli with a Specific Target on the Virulence Factors Controlled by the AggR Master Regulator. Pol J Microbiol 2023; 72:347-354. [PMID: 37875068 PMCID: PMC10725161 DOI: 10.33073/pjm-2023-037] [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: 06/23/2023] [Accepted: 09/08/2023] [Indexed: 10/26/2023] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) strains have been linked to several outbreaks of severe diarrhea around the world, and this bacterium is now commonly resistant to antibiotics. As part of the pathophysiology of EAEC, the characteristic pattern of adherence looks like stacked bricks on the intestinal epithelium. This phenotype depends on an aggregative adhesion plasmid (pAA), which codes for a regulatory protein named AggR. The AggR protein is a master regulator that transcriptionally actives the main virulence genes in this E. coli pathotype, such as those that encode the aggregative adhesion fimbriae, dispersin and its secretion apparatus, Aar regulatory protein, and type VI secretion system. Several reports have shown that AggR positively affects most EAEC virulence genes, functioning as a classic transcriptional activator in the promoter region of these genes, interacting with the RNA polymerase. This minireview article integrates the information about virulence determinants of EAEC controlled by the AggR regulator.
Collapse
Affiliation(s)
| | | | | | - Miguel A. Ares
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, México City, México
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, México
| | | |
Collapse
|
7
|
Mwape K, Bosomprah S, Chibesa K, Silwamba S, Luchen CC, Sukwa N, Mubanga C, Phiri B, Chibuye M, Liswaniso F, Somwe P, Chilyabanyama O, Chisenga CC, Muyoyeta M, Simuyandi M, Barnard TG, Chilengi R. Prevalence of Diarrhoeagenic Escherichia coli among Children Aged between 0-36 Months in Peri-Urban Areas of Lusaka. Microorganisms 2023; 11:2790. [PMID: 38004801 PMCID: PMC10673189 DOI: 10.3390/microorganisms11112790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Diarrhoea is a major contributor to childhood morbidity and mortality in developing countries, with diarrhoeagenic Escherichia coli being among the top aetiological agents. We sought to investigate the burden and describe the diarrhoeagenic E. coli pathotypes causing diarrhoea among children in peri-urban areas of Lusaka, Zambia. This was a facility-based surveillance study conducted over an 8-month period from 2020 to 2021. Stool samples were collected from children aged 0-3 years presenting with diarrhoea at five peri-urban health facilities in Lusaka. Stool samples were tested for diarrhoeagenic E. coli using the Novodiag bacterial GE+® panel, a platform utilising real-time PCR and microarray technology to detect bacterial pathogens. Of the 590 samples tested, diarrhoeagenic E. coli were detected in 471 (76.1%). The top three pathogens were enteropathogenic E. coli 45.4% (n = 268), enteroaggregative E. coli 39.5% (n = 233), and enterotoxigenic E. coli 29.7% (n = 176). Our results revealed that 50.1% of the diarrhoeagenic E. coli positive samples comprised multiple pathotypes of varying virulence gene combinations. Our study demonstrates a high prevalence of diarrhoeagenic E. coli in childhood diarrhoea and the early exposure (<12 months) of children to enteric pathogens. This calls for the early implementation of preventive interventions for paediatric diarrhoea.
Collapse
Affiliation(s)
- Kapambwe Mwape
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
- Water and Health Research Center, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa;
- Department of Basic Medical Sciences, Michael Chilufya Sata School of Medicine, Copperbelt University, Ndola P.O. Box 71191, Zambia
| | - Samuel Bosomprah
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
- Department of Biostatistics, School of Public Health, University of Ghana, Accra P.O. Box LG13, Ghana
| | - Kennedy Chibesa
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, P.O. Box 339, Bloemfontein 9300, South Africa
| | - Suwilanji Silwamba
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka P.O. Box 50110, Zambia
| | - Charlie Chaluma Luchen
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
- Amsterdam Institute of Infection and Immunity, Amsterdam University Medical Centers, 1105 AZ Amsterdam, The Netherlands
- Department of Global Health, Amsterdam Institute for Global Health and Development (AIGHD), Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Nsofwa Sukwa
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
| | - Cynthia Mubanga
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
- Division of Medical Microbiology, Department of Pathology, Stellenbosch University & National Health Laboratory Service, Tygerberg Hospital Francie van Zijl Drive, P.O. Box 241, Cape Town 8000, South Africa
| | - Bernard Phiri
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
| | - Mwelwa Chibuye
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
- Amsterdam Institute of Infection and Immunity, Amsterdam University Medical Centers, 1105 AZ Amsterdam, The Netherlands
- Department of Global Health, Amsterdam Institute for Global Health and Development (AIGHD), Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Fraser Liswaniso
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
| | - Paul Somwe
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
| | - Obvious Chilyabanyama
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
| | - Caroline Cleopatra Chisenga
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
| | - Monde Muyoyeta
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
| | - Michelo Simuyandi
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
| | - Tobias George Barnard
- Water and Health Research Center, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa;
| | - Roma Chilengi
- Enteric Disease and Vaccine Research Unit, Center for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (K.M.); (S.S.); (N.S.); (M.C.); (R.C.)
| |
Collapse
|
8
|
Mohamed MYI, Habib I. Pathogenic E. coli in the Food Chain across the Arab Countries: A Descriptive Review. Foods 2023; 12:3726. [PMID: 37893619 PMCID: PMC10606471 DOI: 10.3390/foods12203726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 09/29/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Foodborne bacterial infections caused by pathogens are a widespread problem in the Middle East, leading to significant economic losses and negative impacts on public health. This review aims to offer insights into the recent literature regarding the occurrence of harmful E. coli bacteria in the food supply of Arab countries. Additionally, it aims to summarize existing information on health issues and the state of resistance to antibiotics. The reviewed evidence highlights a lack of a comprehensive understanding of the extent to which harmful E. coli genes are present in the food supply of Arab countries. Efforts to identify the source of harmful E. coli in the Arab world through molecular characterization are limited. The Gulf Cooperation Council (GCC) countries have conducted few surveys specifically targeting harmful E. coli in the food supply. Despite having qualitative data that indicate the presence or absence of harmful E. coli, there is a noticeable absence of quantitative data regarding the actual numbers of harmful E. coli in chicken meat supplies across all Arab countries. While reports about harmful E. coli in animal-derived foods are common, especially in North African Arab countries, the literature emphasized in this review underscores the ongoing challenge that harmful E. coli pose to food safety and public health in Arab countries.
Collapse
Affiliation(s)
- Mohamed-Yousif Ibrahim Mohamed
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab of Emirates University, Al Ain P.O. Box 1555, United Arab Emirates
| | - Ihab Habib
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab of Emirates University, Al Ain P.O. Box 1555, United Arab Emirates
- Department of Environmental Health, High Institute of Public Health, Alexandria University, Alexandria P.O. Box 21511, Egypt
- ASPIRE Research Institute for Food Security in the Drylands (ARIFSID), United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
9
|
Akinlabi OC, Dada RA, Nwoko ESQA, Okeke IN. PCR diagnostics are insufficient for the detection of Diarrhoeagenic Escherichia coli in Ibadan, Nigeria. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001539. [PMID: 37549136 PMCID: PMC10406320 DOI: 10.1371/journal.pgph.0001539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/10/2023] [Indexed: 08/09/2023]
Abstract
Understanding the contribution of different diarrhoeagenic Escherichia coli pathotypes to disease burden is critical to mapping risk and informing vaccine development. Targeting select virulence genes by PCR is the diagnostic approach of choice in high-burden, least-resourced African settings. We compared the performance of a commonly-used multiplex protocol to whole genome sequencing (WGS). PCR was applied to 3,815 E. coli isolates from 120 children with diarrhoea and 357 healthy controls. Three or more isolates per specimen were also Illumina-sequenced. Following quality assurance, ARIBA and Virulencefinder database were used to identify virulence targets. Root cause analysis of deviant PCR results was performed by examining target sensitivity using BLAST, Sanger sequencing false-positive amplicons, and identifying lineages prone to false-positivity using in-silico multilocus sequence typing and a Single Nucleotide Polymorphism phylogeny constructed using IQTree. The sensitivity and positive predictive value of PCR compared to WGS ranged from 0-77.8% while specificity ranged from 74.5-94.7% for different pathotypes. WGS identified more enteroaggregative E. coli (EAEC), fewer enterotoxigenic E. coli (ETEC) and none of the Shiga toxin-producing E. coli detected by PCR, painting a considerably different epidemiological picture. Use of the CVD432 target resulted in EAEC under-detection, and enteropathogenic E. coli eae primers mismatched more recently described intimin alleles common in our setting. False positive ETEC were over-represented among West Africa-predominant ST8746 complex strains. PCR precision varies with pathogen genome so primers optimized for use in one part of the world may have noticeably lower sensitivity and specificity in settings where different pathogen lineages predominate.
Collapse
Affiliation(s)
- Olabisi C. Akinlabi
- Faculty of Pharmacy, Department of Pharmaceutical Microbiology, University of Ibadan, Oyo, Nigeria
| | - Rotimi A. Dada
- Faculty of Pharmacy, Bowen University Iwo and Department of Pharmaceutical Microbiology, College of Health Sciences, Medical Laboratory Science Programme, Ahmadu Bello University, Zaria, Nigeria
| | - El-shama Q. A. Nwoko
- Faculty of Pharmacy, Department of Pharmaceutical Microbiology, University of Ibadan, Oyo, Nigeria
| | - Iruka N. Okeke
- Faculty of Pharmacy, Department of Pharmaceutical Microbiology, University of Ibadan, Oyo, Nigeria
| |
Collapse
|
10
|
Llorente MT, Escudero R, Ramiro R, Remacha MA, Martínez-Ruiz R, Galán-Sánchez F, de Frutos M, Elía M, Onrubia I, Sánchez S. Enteroaggregative Escherichia coli as etiological agent of endemic diarrhea in Spain: A prospective multicenter prevalence study with molecular characterization of isolates. Front Microbiol 2023; 14:1120285. [PMID: 37065134 PMCID: PMC10100739 DOI: 10.3389/fmicb.2023.1120285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/01/2023] [Indexed: 03/22/2023] Open
Abstract
BackgroundEnteroaggregative Escherichia coli (EAEC) is increasingly associated with domestically acquired diarrheal episodes in high-income countries, particularly among children. However, its specific role in endemic diarrhea in this setting remains under-recognized and information on molecular characteristics of such EAEC strains is limited. We aimed to investigate the occurrence of EAEC in patients with non-travel related diarrhea in Spain and molecularly characterize EAEC strains associated with illness acquired in this high-income setting.MethodsIn a prospective multicenter study, stool samples from diarrheal patients with no history of recent travel abroad (n = 1,769) were collected and processed for detection of EAEC and other diarrheagenic E. coli (DEC) pathotypes by PCR. An additional case–control study was conducted among children ≤5 years old. Whole-genome sequences (WGS) of the resulting EAEC isolates were obtained.ResultsDetection of DEC in the study population. DEC was detected in 23.2% of patients aged from 0 to 102 years, with EAEC being one of the most prevalent pathotypes (7.8%) and found in significantly more patients ≤5 years old (9.8% vs. 3.4%, p < 0.001). Although not statistically significant, EAEC was more frequent in cases than in controls. WGS-derived characterization of EAEC isolates. Sequence type (ST) 34, ST200, ST40, and ST10 were the predominant STs. O126:H27, O111:H21, and O92:H33 were the predominant serogenotypes. Evidence of a known variant of aggregative adherence fimbriae (AAF) was found in 89.2% of isolates, with AAF/V being the most frequent. Ten percent of isolates were additionally classified as presumptive extraintestinal pathogenic E. coli (ExPEC), uropathogenic E. coli (UPEC), or both, and belonged to clonal lineages that could be specifically associated with extraintestinal infections.ConclusionEAEC was the only bacterial enteric pathogen detected in a significant proportion of cases of endemic diarrhea in Spain, especially in children ≤5 years old. In particular, O126:H27-ST200, O111:H21-ST40, and O92:H33-ST34 were the most important subtypes, with all of them infecting both patients and asymptomatic individuals. Apart from this role as an enteric pathogen, a subset of these domestically acquired EAEC strains revealed an additional urinary/systemic pathogenic potential.
Collapse
Affiliation(s)
- María Teresa Llorente
- Reference and Research Laboratory on Food and Waterborne Bacterial Infections, National Center for Microbiology, Institute of Health Carlos III, Madrid, Spain
- Reference and Research Laboratory on Special Pathogens, National Center for Microbiology, Institute of Health Carlos III, Madrid, Spain
| | - Raquel Escudero
- Reference and Research Laboratory on Special Pathogens, National Center for Microbiology, Institute of Health Carlos III, Madrid, Spain
| | - Raquel Ramiro
- Reference and Research Laboratory on Food and Waterborne Bacterial Infections, National Center for Microbiology, Institute of Health Carlos III, Madrid, Spain
| | - María Antonia Remacha
- Servicio de Microbiología Clínica, Complejo Asistencial Universitario de León, León, Spain
| | - Rocío Martínez-Ruiz
- Servicio de Microbiología y Parasitología, Hospital Puerta de Hierro Majadahonda, Majadahonda, Spain
| | | | - Mónica de Frutos
- Servicio de Microbiología, Hospital Universitario del Río Hortega, Valladolid, Spain
| | - Matilde Elía
- Servicio de Microbiología Clínica, Hospital Universitario de Navarra, Pamplona, Spain
| | - Isabel Onrubia
- Pediatría, Centro de Salud Valle de la Oliva, Majadahonda, Spain
| | - Sergio Sánchez
- Reference and Research Laboratory on Food and Waterborne Bacterial Infections, National Center for Microbiology, Institute of Health Carlos III, Madrid, Spain
- *Correspondence: Sergio Sánchez,
| |
Collapse
|
11
|
Osman M, Kassem II, Dabboussi F, Cummings KJ, Hamze M. The indelible toll of enteric pathogens: Prevalence, clinical characterization, and seasonal trends in patients with acute community-acquired diarrhea in disenfranchised communities. PLoS One 2023; 18:e0282844. [PMID: 36913372 PMCID: PMC10010529 DOI: 10.1371/journal.pone.0282844] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/23/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND There is little information on the epidemiology of enteric pathogens in Lebanon, a low- and middle-income country that suffers from a myriad of public health challenges. To address this knowledge gap, we aimed to assess the prevalence of enteric pathogens, identify risk factors and seasonal variations, and describe associations between pathogens among diarrheic patients in the Lebanese community. METHODOLOGY AND PRINCIPAL FINDINGS A multicenter cross-sectional community-based study was conducted in the north of Lebanon. Stool samples were collected from 360 outpatients suffering from acute diarrhea. Based on fecal examination using the BioFire® FilmArray® Gastrointestinal Panel assay, the overall prevalence of enteric infections was 86.1%. Enteroaggregative Escherichia coli (EAEC) was the most frequently identified (41.7%), followed by enteropathogenic E. coli (EPEC) (40.8%) and rotavirus A (27.5%). Notably, two cases of Vibrio cholerae were identified, while Cryptosporidium spp. (6.9%) was the most common parasitic agent. Overall, 27.7% (86/310) of the cases were single infections, and the majority, 73.3% (224/310), were mixed infections. Multivariable logistic regression models showed that enterotoxigenic E. coli (ETEC) and rotavirus A infections were significantly more likely to occur in the fall and winter compared to the summer. Rotavirus A infections significantly decreased with age but increased in patients living in rural areas or suffering from vomiting. We identified strong associations in the co-occurrence of EAEC, EPEC, and ETEC infections and a higher percentage of rotavirus A and norovirus GI/GII infections among EAEC-positive cases. CONCLUSIONS Several of the enteric pathogens reported in this study are not routinely tested in Lebanese clinical laboratories. However, anecdotal evidence suggests that diarrheal diseases are on the rise due to widespread pollution and the deterioration of the economy. Therefore, this study is of paramount importance to identify circulating etiologic agents and prioritize dwindling resources to control them and limit outbreaks in the future.
Collapse
Affiliation(s)
- Marwan Osman
- Cornell Atkinson Center for Sustainability, Cornell University, Ithaca, NY, United States of America
- Department of Public and Ecosystem Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Issmat I. Kassem
- Center for Food Safety and Department of Food Science and Technology, University of Georgia, Griffin, GA, United States of America
| | - Fouad Dabboussi
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School of Sciences and Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Kevin J. Cummings
- Department of Public and Ecosystem Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Monzer Hamze
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School of Sciences and Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| |
Collapse
|
12
|
González-Fernández D, Cousens S, Rizvi A, Chauhadry I, Soofi SB, Bhutta ZA. Infections and nutrient deficiencies during infancy predict impaired growth at 5 years: Findings from the MAL-ED study in Pakistan. Front Nutr 2023; 10:1104654. [PMID: 36875830 PMCID: PMC9982131 DOI: 10.3389/fnut.2023.1104654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/10/2023] [Indexed: 02/19/2023] Open
Abstract
Background Socio-economic, nutritional, and infectious factors have been associated with impaired infant growth, but how the presence of these factors during infancy affects growth around 5 years is not well understood. Methods This secondary analysis of the MAL-ED cohort included 277 children from Pakistan for whom socio-demographic, breastfeeding, complementary foods, illness, nutritional biomarkers, stool pathogens and environmental enteropathy indicators between 0 and 11 months were recorded. We used linear regression models to analyze associations of these indicators with height-for-age (HAZ), weight-for-age (WAZ) and weight-for-height (WLZ) at 54-66 months (~5 years), and Poisson regression with robust standard errors to estimate risk ratios for stunting and underweight ~5 years, controlling for gender, first available weight, and income. Results Among the 237 infants followed longitudinally and evaluated at about 5 years of age, exclusive breastfeeding was short (median = 14 days). Complementary feeding started before 6 months with rice, bread, noodles, or sugary foods. Roots, dairy products, fruits/vegetables, and animal-source foods were provided later than recommended (9-12 months). Anemia (70.9%), deficiencies in iron (22.0%), zinc (80.0%), vitamin A (53.4%) and iodine (13.3%) were common. Most infants (>90%) presented with diarrhea and respiratory infections in their first year. At ~5 years, low WAZ (mean-1.91 ± 0.06) and LAZ (-2.11 ± 0.06) resulted in high prevalence of stunting (55.5%) and underweight (44.4%) but a relatively low rate of wasting (5.5%). While 3.4% had concurrent stunting and wasting ~5 years, 37.8% of children had coexisting stunting and underweight. A higher income and receiving formula or dairy products during infancy were associated with a higher LAZ ~5 years, but infant's history of hospitalizations and more respiratory infections were associated with lower LAZ and higher risk of stunting ~5 years. Infants' intake of commercial baby foods and higher serum-transferrin receptors were associated with higher WAZ and lower risk of underweight ~5 years. Presence of Campylobacter and fecal neopterin >6.8 nmol/L in the first year were associated with increased risk of underweight ~5 years. Conclusion Growth indicators ~5 years were associated with poverty, inappropriate complementary feeding, and infections during the first year of life, which supports the early start of public health interventions for preventing growth delay ~5 years.
Collapse
Affiliation(s)
| | - Simon Cousens
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Arjumand Rizvi
- Center of Excellence in Women and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Imran Chauhadry
- Center of Excellence in Women and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Sajid Bashir Soofi
- Center of Excellence in Women and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Zulfiqar Ahmed Bhutta
- SickKids Centre for Global Child Health, Toronto, ON, Canada.,Center of Excellence in Women and Child Health, The Aga Khan University, Karachi, Pakistan.,Institute for Global Health and Development, The Aga Khan University, London, United Kingdom
| |
Collapse
|
13
|
Hossain S, Mihrshahi S. Exclusive Breastfeeding and Childhood Morbidity: A Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14804. [PMID: 36429518 PMCID: PMC9691199 DOI: 10.3390/ijerph192214804] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 06/16/2023]
Abstract
Globally, diarrheal diseases and acute respiratory infections are the leading causes of morbidity and mortality in children under 5 years old. The benefits of exclusive breastfeeding in reducing the risk of gastrointestinal and respiratory infections are well documented. Optimal breastfeeding may potentially save the lives of about 800,000 children in low-income settings. Despite the evidence, around 63% of infants from birth to 6 months are not exclusively breastfed worldwide. We searched the literature published between 2010 and 2022 in Medline, Embase, and Scopus on the association between exclusive breastfeeding and infectious diseases. We selected and reviewed 70 relevant studies. Our findings expand and confirm the positive association between exclusive breastfeeding and reduced risk of a number of gastrointestinal, respiratory, and other infections in 60 out of 70 studies observed in both low- and high-income settings. Several studies analyzing exclusive breastfeeding duration reported that a longer exclusive breastfeeding duration is protective against many infectious diseases. This review also reported a lack of standardized definition for measuring exclusive breastfeeding in many studies. Overall, the results highlight the benefits of exclusive breastfeeding in many studies and suggests reporting exclusive breastfeeding in future studies using a consistent definition to enable better monitoring of exclusive breastfeeding rates.
Collapse
|
14
|
Freire CA, Rodrigues BO, Elias WP, Abe CM. Adhesin related genes as potential markers for the enteroaggregative Escherichia coli category. Front Cell Infect Microbiol 2022; 12:997208. [DOI: 10.3389/fcimb.2022.997208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) is an important cause of diarrhea in children and adults worldwide. This pathotype is phenotypically characterized by the aggregative-adherence (AA) pattern in HEp-2 cells and genetically associated to the presence of the aatA gene. EAEC pathogenesis relies in different virulence factors. At least, three types of adhesins have been specifically associated with EAEC strains: the five variants of the aggregative adherence fimbriae (AAF), the aggregative forming pilus (AFP) and more recently, a fibrilar adhesin named CS22. Our study aimed to evaluate the presence of AAF, AFP and CS22-related genes among 110 EAEC strains collected from feces of children with diarrhea. The presence of aggR (EAEC virulence regulator) and genes related to AAFs (aggA, aafA, agg3A, agg4A, agg5A and agg3/4C), AFP (afpA1 and afpR) and CS22 (cseA) was detected by PCR, and the adherence patterns were evaluated on HeLa cells. aggR-positive strains comprised 83.6% of the collection; among them, 80.4% carried at least one AAF-related gene and presented the AA pattern. aggA was the most frequent AAF-related gene (28.4% of aggR+ strains). cseA was detected among aggR+ (16.3%) and aggR- strains (22.2%); non-adherent strains or strains presenting AA pattern were observed in both groups. afpR and afpA1 were exclusively detected among aggR- strains (77.8%), most of which (71.4%) also presented AA pattern. Our results indicate that AAF- and AFP-related genes may contribute to identify EAEC strains, while the presence of cseA and its importance as an EAEC virulence factor and genotypic marker needs to be further evaluated.
Collapse
|
15
|
Rogawski McQuade ET, Brennhofer SA, Elwood SE, McMurry TL, Lewnard JA, Mduma ER, Shrestha S, Iqbal N, Bessong PO, Kang G, Kosek M, Lima AAM, Ahmed T, Liu J, Houpt ER, Platts-Mills JA. Frequency of bystander exposure to antibiotics for enteropathogenic bacteria among young children in low-resource settings. Proc Natl Acad Sci U S A 2022; 119:e2208972119. [PMID: 36037372 PMCID: PMC9457395 DOI: 10.1073/pnas.2208972119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/01/2022] [Indexed: 11/24/2022] Open
Abstract
Children in low-resource settings carry enteric pathogens asymptomatically and are frequently treated with antibiotics, resulting in opportunities for pathogens to be exposed to antibiotics when not the target of treatment (i.e., bystander exposure). We quantified the frequency of bystander antibiotic exposures for enteric pathogens and estimated associations with resistance among children in eight low-resource settings. We analyzed 15,697 antibiotic courses from 1,715 children aged 0 to 2 y from the MAL-ED birth cohort. We calculated the incidence of bystander exposures and attributed exposures to respiratory and diarrheal illnesses. We associated bystander exposure with phenotypic susceptibility of E. coli isolates in the 30 d following exposure and at the level of the study site. There were 744.1 subclinical pathogen exposures to antibiotics per 100 child-years. Enteroaggregative Escherichia coli was the most frequently exposed pathogen, with 229.6 exposures per 100 child-years. Almost all antibiotic exposures for Campylobacter (98.8%), enterotoxigenic E. coli (95.6%), and typical enteropathogenic E. coli (99.4%), and the majority for Shigella (77.6%), occurred when the pathogens were not the target of treatment. Respiratory infections accounted for half (49.9%) and diarrheal illnesses accounted for one-fourth (24.6%) of subclinical enteric bacteria exposures to antibiotics. Bystander exposure of E. coli to class-specific antibiotics was associated with the prevalence of phenotypic resistance at the community level. Antimicrobial stewardship and illness-prevention interventions among children in low-resource settings would have a large ancillary benefit of reducing bystander selection that may contribute to antimicrobial resistance.
Collapse
Affiliation(s)
| | - Stephanie A. Brennhofer
- Division of Infectious Diseases & International Health, University of Virginia, Charlottesville, VA 22908
| | - Sarah E. Elwood
- Division of Infectious Diseases & International Health, University of Virginia, Charlottesville, VA 22908
| | - Timothy L. McMurry
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA 22908
| | - Joseph A. Lewnard
- Division of Epidemiology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720
| | - Estomih R. Mduma
- Haydom Global Health Research Center, Haydom Lutheran Hospital, Haydom, Tanzania
| | - Sanjaya Shrestha
- Walter Reed/AFRIMS Research Unit, Nepal, Kathmandu, 44600, Nepal
| | | | - Pascal O. Bessong
- University of Venda, Thohoyandou, 0950, South Africa
- Center for Global Health Equity, University of Virginia, Charlottesville, VA 22908
| | | | - Margaret Kosek
- Division of Infectious Diseases & International Health, University of Virginia, Charlottesville, VA 22908
- Asociación Benéfica PRISMA, Iquitos, 15088, Peru
| | | | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, 1212, Bangladesh
| | - Jie Liu
- School of Public Health, Qingdao University, Qingdao, Shandong, 266071, China
| | - Eric R. Houpt
- Division of Infectious Diseases & International Health, University of Virginia, Charlottesville, VA 22908
| | - James A. Platts-Mills
- Division of Infectious Diseases & International Health, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
16
|
Agoti CN, Curran MD, Murunga N, Ngari M, Muthumbi E, Lambisia AW, Frost SDW, Blacklaws BA, Nokes DJ, Drumright LN. Differences in epidemiology of enteropathogens in children pre- and post-rotavirus vaccine introduction in Kilifi, coastal Kenya. Gut Pathog 2022; 14:32. [PMID: 35915480 PMCID: PMC9340678 DOI: 10.1186/s13099-022-00506-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/18/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Kenya introduced Rotarix® (GlaxoSmithKline Biologicals, Rixensart, Belgium) vaccination into its national immunization programme beginning July 2014. The impact of this vaccination program on the local epidemiology of various known enteropathogens is not fully understood. METHODS We used a custom TaqMan Array Card (TAC) to screen for 28 different enteropathogens in 718 stools from children aged less than 13 years admitted to Kilifi County Hospital, coastal Kenya, following presentation with diarrhea in 2013 (before vaccine introduction) and in 2016-2018 (after vaccine introduction). Pathogen positivity rate differences between pre- and post-Rotarix® vaccination introduction were examined using both univariate and multivariable logistic regression models. RESULTS In 665 specimens (92.6%), one or more enteropathogen was detected, while in 323 specimens (48.6%) three or more enteropathogens were detected. The top six detected enteropathogens were: enteroaggregative Escherichia coli (EAggEC; 42.1%), enteropathogenic Escherichia coli (EPEC; 30.2%), enterovirus (26.9%), rotavirus group A (RVA; 24.8%), parechovirus (16.6%) and norovirus GI/GII (14.4%). Post-rotavirus vaccine introduction, there was a significant increase in the proportion of samples testing positive for EAggEC (35.7% vs. 45.3%, p = 0.014), cytomegalovirus (4.2% vs. 9.9%, p = 0.008), Vibrio cholerae (0.0% vs. 2.3%, p = 0.019), Strongyloides species (0.8% vs. 3.6%, p = 0.048) and Dientamoeba fragilis (2.1% vs. 7.8%, p = 0.004). Although not reaching statistical significance, the positivity rate of adenovirus 40/41 (5.8% vs. 7.3%, p = 0.444), norovirus GI/GII (11.2% vs. 15.9%, p = 0.089), Shigella species (8.7% vs. 13.0%, p = 0.092) and Cryptosporidium spp. (11.6% vs. 14.7%, p = 0.261) appeared to increase post-vaccine introduction. Conversely, the positivity rate of sapovirus decreased significantly post-vaccine introduction (7.8% vs. 4.0%, p = 0.030) while that of RVA appeared not to change (27.4% vs. 23.5%, p = 0.253). More enteropathogen coinfections were detected per child post-vaccine introduction compared to before (mean: 2.7 vs. 2.3; p = 0.0025). CONCLUSIONS In this rural Coastal Kenya setting, childhood enteropathogen infection burden was high both pre- and post-rotavirus vaccination introduction. Children who had diarrheal admissions post-vaccination showed an increase in coinfections and changes in specific enteropathogen positivity rates. This study highlights the utility of multipathogen detection platforms such as TAC in understanding etiology of childhood acute gastroenteritis in resource-limited regions.
Collapse
Affiliation(s)
- Charles N Agoti
- Epidemiology and Demography Department, Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, P.O. Box 230, Kilifi, 80108, Kenya.
- School of Health and Human Sciences, Pwani University, Kilifi, Kenya.
| | - Martin D Curran
- Public Health England, Cambridge, UK
- Clinical Microbiology and Public Health Laboratory, Addenbrooke's Hospital, Cambridge, UK
| | - Nickson Murunga
- Epidemiology and Demography Department, Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Moses Ngari
- Epidemiology and Demography Department, Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Esther Muthumbi
- Epidemiology and Demography Department, Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Arnold W Lambisia
- Epidemiology and Demography Department, Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Simon D W Frost
- Microsoft Research, Building 99, 14820 NE 36th St., Redmond, WA 98052, USA
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Public Health, London School of Hygiene and Tropical Medicine, Keppel St, Bloomsbury, London, WC1E 7HT, UK
| | | | - D James Nokes
- Epidemiology and Demography Department, Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, P.O. Box 230, Kilifi, 80108, Kenya
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Lydia N Drumright
- Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Washington, Washington, USA
| |
Collapse
|
17
|
Lambrecht NJ, Bridges D, Wilson ML, Adu B, Eisenberg JNS, Folson G, Baylin A, Jones AD. Associations of bacterial enteropathogens with systemic inflammation, iron deficiency, and anemia in preschool-age children in southern Ghana. PLoS One 2022; 17:e0271099. [PMID: 35802561 PMCID: PMC9269377 DOI: 10.1371/journal.pone.0271099] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 06/23/2022] [Indexed: 11/19/2022] Open
Abstract
Anemia remains a pervasive public health problem among preschool-age children in Ghana. Recent analyses have found that anemia in Ghanaian children, particularly in Southern regions, is largely attributable to infectious causes, rather than nutritional factors. Infections with enteropathogens can reduce iron absorption and increase systemic inflammation, but few studies have examined direct links between enteropathogens and anemia. This study investigated associations between detection of individual bacterial enteropathogens and systemic inflammation, iron deficiency, and anemia among 6- to 59-month-old children in Greater Accra, Ghana. Serum samples were analyzed from a cross-sectional sample of 262 children for concentrations of hemoglobin (Hb), biomarkers of systemic inflammation [C-reactive protein (CRP) and α-1-acid glycoprotein (AGP)], and biomarkers of iron status [serum ferritin (SF) and serum transferrin receptor (sTfR)]. Stool samples were analyzed for ten bacterial enteropathogens using qPCR. We estimated associations between presence of each enteropathogen and elevated systemic inflammation (CRP > 5 mg/L and AGP > 1 g/L), iron deficiency (SF < 12 μg/L and sTfR > 8.3 mg/L) and anemia (Hb < 110 g/L). Enteropathogens were detected in 87% of children’s stool despite a low prevalence of diarrhea (6.5%). Almost half (46%) of children had anemia while one-quarter (24%) had iron deficiency (low SF). Despite finding no associations with illness symptoms, Campylobacter jejuni/coli detection was strongly associated with elevated CRP [Odds Ratio (95% CI): 3.49 (1.45, 8.41)] and elevated AGP [4.27 (1.85, 9.84)]. Of the pathogens examined, only enteroinvasive Escherichia coli/Shigella spp. (EIEC/Shigella) was associated with iron deficiency, and enteroaggregative Escherichia coli (EAEC) [1.69 (1.01, 2.84)] and EIEC/Shigella [2.34 (1.15, 4.76)] were associated with anemia. These results suggest that certain enteroinvasive pathogenic bacteria may contribute to child anemia. Reducing exposure to enteropathogens through improved water, sanitation, and hygiene practices may help reduce the burden of anemia in young Ghanaian children.
Collapse
Affiliation(s)
- Nathalie J. Lambrecht
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
- Institute of Public Health, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Research Department 2, Potsdam Institute for Climate Impact Research (PIK), Member of the Leibniz Association, Potsdam, Germany
- * E-mail:
| | - Dave Bridges
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Mark L. Wilson
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Bright Adu
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Joseph N. S. Eisenberg
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Gloria Folson
- Department of Nutrition, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Ana Baylin
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Andrew D. Jones
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
18
|
Collard JM, Andrianonimiadana L, Habib A, Rakotondrainipiana M, Andriantsalama P, Randriamparany R, Rabenandrasana MAN, Weill FX, Sauvonnet N, Randremanana RV, Guillemot V, Vonaesch P, Sansonetti PJ. High prevalence of small intestine bacteria overgrowth and asymptomatic carriage of enteric pathogens in stunted children in Antananarivo, Madagascar. PLoS Negl Trop Dis 2022; 16:e0009849. [PMID: 35533199 PMCID: PMC9119516 DOI: 10.1371/journal.pntd.0009849] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 05/19/2022] [Accepted: 04/01/2022] [Indexed: 11/26/2022] Open
Abstract
Environmental Enteric Dysfunction (EED) refers to an incompletely defined syndrome of inflammation, reduced absorptive capacity, and reduced barrier function in the small intestine. It is widespread among children and adults in low- and middle-income countries and is also associated with poor sanitation and certain gut infections possibly resulting in an abnormal gut microbiota, small intestinal bacterial overgrowth (SIBO) and stunting. We investigated bacterial pathogen exposure in stunted and non-stunted children in Antananarivo, Madagascar by collecting fecal samples from 464 children (96 severely stunted, 104 moderately stunted and 264 non-stunted) and the prevalence of SIBO in 109 duodenal aspirates from stunted children (61 from severely stunted and 48 from moderately stunted children). SIBO assessed by both aerobic and anaerobic plating techniques was very high: 85.3% when selecting a threshold of ≥105 CFU/ml of bacteria in the upper intestinal aspirates. Moreover, 58.7% of the children showed more than 106 bacteria/ml in these aspirates. The most prevalent cultivated genera recovered were Streptococcus, Neisseria, Staphylococcus, Rothia, Haemophilus, Pantoea and Branhamella. Feces screening by qPCR showed a high prevalence of bacterial enteropathogens, especially those categorized as being enteroinvasive or causing mucosal disruption, such as Shigella spp., enterotoxigenic Escherichia coli, enteropathogenic E. coli and enteroaggregative E. coli. These pathogens were detected at a similar rate in stunted children and controls, all showing no sign of severe diarrhea the day of inclusion but both living in a highly contaminated environment (slum-dwelling). Interestingly Shigella spp. was the most prevalent enteropathogen found in this study (83.3%) without overrepresentation in stunted children. About 2 million children under the age of 5 suffer from stunted growth in Madagascar. Although deficient diet is the major cause of undernutrition, impaired absorption or assimilation caused by Environmental Enteric dysfunction (EED) has been proposed to play an important role in stunting. EED is widespread among children and adults in low- and middle-income countries (LMIC) and is also associated with undernutrition, poor sanitation, certain gut infections resulting in an abnormal gut microbiota and small intestinal bacterial overgrowth (SIBO) although the role of SIBO in EED remains unclear. The current study highlights the presence at high concentrations of bacterial taxa usually found in the oro-pharyngeal sphere in a high number of duodenal fluids of stunted children. This uncommon presence suggests a decompartmentalization of the gastrointestinal tract and a possible pro-inflammatory effect due to the ectopic presence of some of these bacteria in the duodenum. The study also points to a high prevalence of enteropathogens (especially Shigella spp.) in the feces of both stunted and control children, hence preventing from proposing a direct association with stunting. This suggests that, beside combatting poverty and improving diet, environmental sanitation, quality of water sources, hygiene promotion and health education are key points to mitigate stunting and restore nutritional benefits.
Collapse
Affiliation(s)
- Jean-Marc Collard
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, Antananarivo, Madagascar
- * E-mail:
| | - Lova Andrianonimiadana
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | - Azimdine Habib
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | | | - Prisca Andriantsalama
- Unité d’Epidémiologie et de Recherche Clinique, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | - Ravaka Randriamparany
- Unité d’Epidémiologie et de Recherche Clinique, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | - M. A. N. Rabenandrasana
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | - François-Xavier Weill
- Centre National de Référence des Escherichia coli, Shigella et Salmonella, Unité des Bactéries Pathogènes Entériques, Institut Pasteur, Paris, France
| | - Nathalie Sauvonnet
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France
| | | | - Vincent Guillemot
- Hub of Bioinformatics and Biostatistics, Institut Pasteur, Paris, France
| | - Pascale Vonaesch
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France
| | | | | |
Collapse
|
19
|
Site specific incidence rate of genomic subtypes of enteropathogenic Escherichia coli and association with enteric inflammation and child growth. Sci Rep 2022; 12:5724. [PMID: 35388098 PMCID: PMC8986875 DOI: 10.1038/s41598-022-09730-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
There is a lack of information highlighting the possible association between the genomic subtypes of enteropathogenic Escherichia coli (EPEC) on environmental enteric dysfunction (EED) and on linear growth during childhood. Genomic subtypes of EPEC from stool samples collected from 1705 children enrolled in the MAL-ED birth cohort were detected by TaqMan Array Cards. We measured site-specific incidence rate by using Poisson regression models, identified the risk factors and estimated the association of genomic subtypes of EPEC with the composite EED score and linear growth at 24 months of age. In general, the highest incidence rate (39%) was found among children having aEPEC infection, which was the greatest in Tanzania (54%). Exclusive breastfeeding and having an improved sanitation facility were found to be protective factors against EPEC infection. In the multivariate models, in overall effect after adjusting for the potential covariates aEPEC showed strong positive associations with the EED scores and tEPEC showed a positive association with poor linear growth at 24 months of age. Our analyses may lay the cornerstone for a prospective epidemiologic investigation for a potential vaccine development aimed at reducing the burden of EPEC infections and combat childhood malnutrition.
Collapse
|
20
|
Palit P, Das R, Haque MA, Hasan MM, Noor Z, Mahfuz M, Faruque ASG, Ahmed T. Risk Factors for Norovirus Infections and Their Association with Childhood Growth: Findings from a Multi-Country Birth Cohort Study. Viruses 2022; 14:v14030647. [PMID: 35337054 PMCID: PMC8954848 DOI: 10.3390/v14030647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/05/2022] [Accepted: 03/18/2022] [Indexed: 12/13/2022] Open
Abstract
The prevalence of norovirus infections in different geographical locations and their attribution to childhood diarrhea is well established. However, there are no reports showing possible relationships of different norovirus genogroups with subsequent childhood malnutrition. In this study, we attempted to establish a potential association between asymptomatic norovirus infections with childhood growth faltering during. Non-diarrheal stools were collected from 1715 children enrolled in locations in a multi-county birth cohort study across eight different geographical locations and were assessed for norovirus genogroup I (GI) and norovirus genogroup II (GII). Asymptomatic norovirus GI infections were negatively associated with monthly length-for-age Z score/LAZ (β = -0.53, 95% CI: -0.73, -0.50) and weight-for-age Z score/WAZ (β = -0.39, 95% CI: -0.49, -0.28), respectively. The burden of asymptomatic norovirus GI infections was negatively associated with LAZ (β = -0.46, 95% CI: -0.67, -0.41) and WAZ (β = -0.66, 95% CI: -0.86, -0.53) at 2 years of age, whilst the burden of asymptomatic norovirus GII infections was negatively associated with WAZ (β = -0.27, 95% CI: -0.45, -0.25) at 2 years of age. Our findings warrant acceleration in attempts to develop vaccines against norovirus GI and norovirus GII, with the aim of minimizing the long-term sequelae on childhood growth.
Collapse
Affiliation(s)
- Parag Palit
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (P.P.); (R.D.); (M.M.H.); (M.M.); (A.S.G.F.); (T.A.)
| | - Rina Das
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (P.P.); (R.D.); (M.M.H.); (M.M.); (A.S.G.F.); (T.A.)
| | - Md. Ahshanul Haque
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (P.P.); (R.D.); (M.M.H.); (M.M.); (A.S.G.F.); (T.A.)
- Correspondence:
| | - Md. Mehedi Hasan
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (P.P.); (R.D.); (M.M.H.); (M.M.); (A.S.G.F.); (T.A.)
| | - Zannatun Noor
- Emerging Infections and Parasitology Laboratory, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh;
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (P.P.); (R.D.); (M.M.H.); (M.M.); (A.S.G.F.); (T.A.)
- Faculty of Medicine and Life Sciences, University of Tampere, 33100 Tampere, Finland
| | - Abu Syed Golam Faruque
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (P.P.); (R.D.); (M.M.H.); (M.M.); (A.S.G.F.); (T.A.)
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (P.P.); (R.D.); (M.M.H.); (M.M.); (A.S.G.F.); (T.A.)
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
21
|
McCormick BJJ, Richard SA, Murray-Kolb LE, Kang G, Lima AAM, Mduma E, Kosek MN, Rogawski McQuade ET, Houpt ER, Bessong P, Shrestha S, Bhutta Z, Ahmed T, Caulfield LE. Full breastfeeding protection against common enteric bacteria and viruses: results from the MAL-ED cohort study. Am J Clin Nutr 2022; 115:759-769. [PMID: 34849524 PMCID: PMC8895209 DOI: 10.1093/ajcn/nqab391] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/19/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Breastfeeding is known to reduce the risk of enteropathogen infections, but protection from specific enteropathogens is not well characterized. OBJECTIVE The aim was to estimate the association between full breastfeeding (days fed breast milk exclusively or with nonnutritive liquids) and enteropathogen detection. METHODS A total of 2145 newborns were enrolled at 8 sites, of whom 1712 had breastfeeding and key enteropathogen data through 6 mo. We focused on 11 enteropathogens: adenovirus 40/41, norovirus, sapovirus, astrovirus, and rotavirus, enterotoxigenic Escherichia coli (ETEC), Campylobacter spp., and typical enteropathogenic E. coli as well as entero-aggregative E. coli, Shigella and Cryptosporidium. Logistic regression was used to estimate the risk of enteropathogen detection in stools and survival analysis was used to estimate the timing of first detection of an enteropathogen. RESULTS Infants with 10% more days of full breastfeeding within the preceding 30 d of a stool sample were less likely to have the 3 E. coli and Campylobacter spp. detected in their stool (mean odds: 0.92-0.99) but equally likely (0.99-1.02) to have the viral pathogens detected in their stool. A 10% longer period of full breastfeeding from birth was associated with later first detection of the 3 E. coli, Campylobacter, adenovirus, astrovirus, and rotavirus (mean HRs of 0.52-0.75). The hazards declined and point estimates were not statistically significant at 3 mo. CONCLUSIONS In this large multicenter cohort study, full breastfeeding was associated with lower likelihood of detecting 4 important enteric pathogens in the first 6 mo of life. These results also show that full breastfeeding is related to delays in the first detection of some bacterial and viral pathogens in the stool. As several of these pathogens are risk factors for poor growth during childhood, this work underscores the importance of exclusive or full breastfeeding during the first 6 mo of life to optimize early health.
Collapse
Affiliation(s)
| | - Stephanie A Richard
- Fogarty International Center/National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | - Eric R Houpt
- University of Virginia, Charlottesville, VA, USA
| | | | | | | | | | - Laura E Caulfield
- The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | |
Collapse
|
22
|
Waltmann A, McQuade ETR, Chinkhumba J, Operario DJ, Mzembe E, Itoh M, Kayange M, Puerto-Meredith SM, Mathanga DP, Juliano JJ, Carroll I, Bartelt LA, Gutman JR, Meshnick SR. The positive effect of malaria IPTp-SP on birthweight is mediated by gestational weight gain but modifiable by maternal carriage of enteric pathogens. EBioMedicine 2022; 77:103871. [PMID: 35217408 PMCID: PMC8866062 DOI: 10.1016/j.ebiom.2022.103871] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Poor pregnancy and birth outcomes are common in sub-Saharan Africa and have complex aetiologies. Sulfadoxine-pyrimethamine (SP), given for intermittent preventive therapy of malaria in pregnancy (IPTp), is one of few existing interventions that improves outcomes of both mother and baby despite widespread SP-resistant malaria. Compelling evidence exists that malaria-independent pathways contribute to this protective effect, but the exact sources of non anti-malarial protection remained unknown. We hypothesized that the beneficial effect of SP on birthweight is mediated by SP activity on maternal factors, including increased gestational weight gain and antibiotic activity on pathogens in the maternal gut. METHODS Expectant mothers from a larger randomized control trial comparing the efficacy of IPTp-SP to IPTp with dihydroartemisinin-piperaquine (DP) were also enrolled in this sub-study study at their first antenatal care visit before commencement of IPTp (n = 105). Participants were followed monthly until delivery. Weights and mid-to-upper-arm circumferences (MUAC) were recorded. Monthly stool samples were collected and screened for five Escherichia coli pathotypes, Shigella spp., Vibrio cholerae, Salmonella, Campylobacter coli/jejuni, and three protozoa (Giardia spp., Entameba histolytica, and Cryptosporidium spp.) using previously validated molecular assays. FINDINGS IPTp-SP vs. IPTP-DP was associated with higher maternal gestational weight gain (GWG) and nutritional indicators (MUAC and body-mass index, BMI). GWG was found to be a mediator of the birthweight and IPTp-SP relationship, as the birthweight of SP infants, but not DP infants, varied according to maternal GWG. The burden of maternal enteric infections was high. The three most commonly observed pathogens were enteroaggregative E. coli (EAEC), atypical enteropathogenic E.coli/enterohaemorrhagic E. coli (aEPEC/EHEC), and typical enteropathogenic E.coli (tEPEC). We found that SP reduced the prevalence of EAEC in a dose-dependent manner. After 3 or more doses, SP-recipients were 90% less likely to be infected with EAEC compared to DP-recipients (ORadj = 0.07, CI95 = 0.12, 0.39, p = 0.002). Compared to DP, this coincided with higher maternal gestational weight gain (GWG) and nutritional indicators (MUAC and body-mass index, BMI). The beneficial effect of SP on maternal GWG, MUAC and BMI, was lower if SP mothers had detectable EAEC, aEPEC/EHEC, tEPEC, and LT-ETEC at baseline. Maternal EAEC and tEPEC at baseline associated with lower birthweight for babies of both SP mothers and DP mothers. When comparing IPTp regimens, the positive effect of SP on birthweight compared to DP was only observed for infants of women who did not test positive for EAEC at baseline (adjusted mean birthweight difference SP vs. DP = 156.0 g, CI95 = -18.0 g, 336.9 g, p = 0.087), though confidence intervals crossed the null. INTERPRETATION Our findings indicate that in pregnant Malawian women, IPTp-SP vs. IPTp-DP is consistently associated with higher MUAC, BMI, and GWG following the WHO-recommended regimen of at least 3 doses, but carriage of maternal gut pathogens before initiation of IPTp lessens this effect. Because GWG was a mediator of the association between birthweight and SP, we show that SP's previously proven positive effect on birthweight is by promoting maternal weight gain. Overall, our results present one plausible pathway SP exerts malaria-independent protection against poor birth outcomes in the context of its waning antimalarial activity and warrants further investigation. FUNDING A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.
Collapse
Affiliation(s)
- Andreea Waltmann
- Institute for Global Health and Infectious Diseases, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| | | | - Jobiba Chinkhumba
- Malaria Alert Centre (MAC), University of Malawi College of Medicine, Blantyre, Malawi
| | - Darwin J Operario
- Division of Infectious Diseases & International Health, Department of Medicine, University of Virginia, VA, USA
| | - Enala Mzembe
- Malaria Alert Centre (MAC), University of Malawi College of Medicine, Blantyre, Malawi
| | - Megumi Itoh
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | - Don P Mathanga
- Malaria Alert Centre (MAC), University of Malawi College of Medicine, Blantyre, Malawi
| | - Jonathan J Juliano
- Institute for Global Health and Infectious Diseases, School of Medicine, University of North Carolina, Chapel Hill, NC, USA; Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Ian Carroll
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Luther A Bartelt
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Julie R Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Steven R Meshnick
- Institute for Global Health and Infectious Diseases, School of Medicine, University of North Carolina, Chapel Hill, NC, USA; Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
23
|
Palit P, Das R, Haque MA, Nuzhat S, Khan SS, Siddiqua TJ, Mahfuz M, Faruque ASG, Ahmed T. Risk Factors for Enterotoxigenic Bacteroides fragilis Infection and Association with Environmental Enteric Dysfunction and Linear Growth in Children: Results from the MAL-ED Study. Am J Trop Med Hyg 2022; 106:915-922. [PMID: 35100563 PMCID: PMC8922507 DOI: 10.4269/ajtmh.21-0780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/27/2021] [Indexed: 01/09/2023] Open
Abstract
Despite reports of enterotoxigenic Bacteroides fragilis (ETBF) isolation from asymptomatic children, no reports exist regarding the possible association of ETBF with long-term complications such as development of environmental enteric dysfunction (EED) and subsequent linear growth faltering in childhood. We aimed to establish a potential association between the burden of asymptomatic ETBF infection and EED and linear growth at 24 months of age using the data collected from 1,715 children enrolled in the multi-country birth cohort study, known as the Etiology, Risk Factors, and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health study. Using Poisson regression models, we evaluated the site-specific incidence rate and, subsequently, identified the risk factors and assessed the association between the burden of ETBF infection and EED score and linear growth at 24 months of age. The overall incidence rate of ETBF infections per 100 child-months across all study sites was 10.6%, with the highest and lowest incidence of ETBF infections being reported in Tanzania (19.6%) and Peru (3.6%), respectively. Female gender, longer duration of breastfeeding, and improved water access, sanitation, and hygiene practices, such as improved drinking water source, improved sanitation, and improved floor material in households, along with enhanced maternal education and less crowding in the households were found to be protective against incidences of ETBF infection. The burden of ETBF infections was found to have significant associations with EED and linear growth faltering at 24 months of age across all the study sites. Our findings warrant regular clinical monitoring to reduce the burden of ETBF infections and diminish the burden of enteropathy and linear growth faltering in childhood.
Collapse
Affiliation(s)
- Parag Palit
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Rina Das
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Md Ahshanul Haque
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Sharika Nuzhat
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Shaila Sharmeen Khan
- Emerging Infections and Parasitology Laboratory, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Towfida Jahan Siddiqua
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh.,Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Abu Syed Golam Faruque
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh.,James P. Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| |
Collapse
|
24
|
Moya-Alvarez V, Sansonetti PJ. Understanding the pathways leading to gut dysbiosis and enteric environmental dysfunction in infants: the influence of maternal dysbiosis and other microbiota determinants during early life. FEMS Microbiol Rev 2022; 46:6516326. [PMID: 35088084 DOI: 10.1093/femsre/fuac004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/10/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Maternal environmental enteric dysfunction (EED) encompasses undernutrition with an inflammatory gut profile, a variable degree of dysbiosis and increased translocation of pathogens in the gut mucosa. Even though recent research findings have shed light on the pathological pathways underlying the establishment of the infant gut dysbiosis, evidence on how maternal EED influences the development of gut dysbiosis and EED in the offspring remains elusive. This review summarizes the current knowledge on the effect of maternal dysbiosis and EED on infant health, and explores recent progress in unraveling the mechanisms of acquisition of a dysbiotic gut microbiota in the offspring. In Western communities, maternal inoculum, delivery mode, perinatal antibiotics, feeding practices, and infections are the major drivers of the infant gut microbiota during the first two years of life. In other latitudes, the infectious burden and maternal malnutrition might introduce further risk factors for infant gut dysbiosis. Novel tools, such as transcriptomics and metabolomics, have become indispensable to analyze the metabolic environment of the infant in utero and post-partum. Human-milk oligosaccharides have essential prebiotic, antimicrobial, and anti-biofilm properties that might offer additional therapeutic opportunities.
Collapse
Affiliation(s)
- Violeta Moya-Alvarez
- Molecular Microbial Pathogenesis - INSERM U1202, Department of Cell Biology and Infection, 28 rue du Dr. Roux, Institut Pasteur, 75015 Paris, France.,Epidemiology of Emergent Diseases Unit, Global Health Department, 25 rue du Dr. Roux, Institut Pasteur, 75015 Paris, France
| | - Philippe J Sansonetti
- Molecular Microbial Pathogenesis - INSERM U1202, Department of Cell Biology and Infection, 28 rue du Dr. Roux, Institut Pasteur, 75015 Paris, France.,Chaire de Microbiologie et Maladies Infectieuses, Collège de France, Paris, France.,The Center for Microbes, Development and Health, Institut Pasteur de Shanghai, China
| |
Collapse
|
25
|
Diarrhea, Dysbiosis, Dysfunction, and the Disastrous Global Health Consequences: Piecing the Puzzle Together. Am J Gastroenterol 2022; 117:98-99. [PMID: 34850781 DOI: 10.14309/ajg.0000000000001555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022]
Abstract
The burden of diarrheal infections globally, including the chronic health consequences, is an important problem. Herein we describe a recent paper published by the Journal and describe how it fits within and advances our knowledge in this area.
Collapse
|
26
|
Wallenborn JT, Vonaesch P. OUP accepted manuscript. Gastroenterol Rep (Oxf) 2022; 10:goac010. [PMID: 35419206 PMCID: PMC8996373 DOI: 10.1093/gastro/goac010] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/10/2021] [Accepted: 02/16/2022] [Indexed: 11/15/2022] Open
Abstract
The intestinal microbiota plays a crucial role in health and changes in its composition are linked with major global human diseases. Fully understanding what shapes the human intestinal microbiota composition and knowing ways of modulating the composition are critical for promotion of life-course health, combating diseases, and reducing global health disparities. We aim to provide a foundation for understanding what shapes the human intestinal microbiota on an individual and global scale, and how interventions could utilize this information to promote life-course health and reduce global health disparities. We briefly review experiences within the first 1,000 days of life and how long-term exposures to environmental elements or geographic specific cultures have lasting impacts on the intestinal microbiota. We also discuss major public health threats linked to the intestinal microbiota, including antimicrobial resistance and disappearing microbial diversity due to globalization. In order to promote global health, we argue that the interplay of the larger ecosystem with intestinal microbiota research should be utilized for future research and urge for global efforts to conserve microbial diversity.
Collapse
Affiliation(s)
- Jordyn T Wallenborn
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Pascale Vonaesch
- Department of Fundamental Microbiology, University of Lausanne, Bâtiment Biophore Campus UNIL-Sorge, Lausanne, Switzerland
- Corresponding author. Department of Fundamental Microbiology, University of Lausanne, 1015 Lausanne, Switzerland. Tel: +41-21-692-5600;
| |
Collapse
|
27
|
Site specific incidence rate of virulence related genes of enteroaggregative Escherichia coli and association with enteric inflammation and growth in children. Sci Rep 2021; 11:23178. [PMID: 34848801 PMCID: PMC8632913 DOI: 10.1038/s41598-021-02626-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/15/2021] [Indexed: 01/19/2023] Open
Abstract
There is a lack of information highlighting the possible association between strain carrying genes of enteroaggregative Escherichia coli (EAEC) and environmental enteric dysfunction (EED) and on linear growth during childhood. Strain carrying genes of EAEC from stool samples collected from 1705 children enrolled in the MAL-ED birth cohort were detected by TaqMan Array Cards. We measured site-specific incidence rate by using Poisson regression models, identified the risk factors and estimated the associations of strain carrying genes of EAEC with the composite EED score and linear growth at 24 months of age. Overall highest incidence rate (43.3%) was found among children having infection with the aggR gene, which was the greatest in Tanzania (56.7%). Low maternal education, lack of improved floor, and ownership of domestic cattle were found to be risk factors for EAEC infection. In the multivariate models, after adjusting the potential covariates, strain carrying genes of EAEC showed strong positive associations with the EED scores and with poor linear growth at 24 months of age. Our analyses may lay the cornerstone for a prospective epidemiologic investigation for a potential vaccine development aimed at reducing the burden of EAEC infections and combat childhood malnutrition.
Collapse
|
28
|
Huus KE, Hoang TT, Creus-Cuadros A, Cirstea M, Vogt SL, Knuff-Janzen K, Sansonetti PJ, Vonaesch P, Finlay BB. Cross-feeding between intestinal pathobionts promotes their overgrowth during undernutrition. Nat Commun 2021; 12:6860. [PMID: 34824233 PMCID: PMC8617199 DOI: 10.1038/s41467-021-27191-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 11/08/2021] [Indexed: 12/18/2022] Open
Abstract
Child undernutrition is a global health issue associated with a high burden of infectious disease. Undernourished children display an overabundance of intestinal pathogens and pathobionts, and these bacteria induce enteric dysfunction in undernourished mice; however, the cause of their overgrowth remains poorly defined. Here, we show that disease-inducing human isolates of Enterobacteriaceae and Bacteroidales spp. are capable of multi-species symbiotic cross-feeding, resulting in synergistic growth of a mixed community in vitro. Growth synergy occurs uniquely under malnourished conditions limited in protein and iron: in this context, Bacteroidales spp. liberate diet- and mucin-derived sugars and Enterobacteriaceae spp. enhance the bioavailability of iron. Analysis of human microbiota datasets reveals that Bacteroidaceae and Enterobacteriaceae are strongly correlated in undernourished children, but not in adequately nourished children, consistent with a diet-dependent growth synergy in the human gut. Together these data suggest that dietary cross-feeding fuels the overgrowth of pathobionts in undernutrition.
Collapse
Affiliation(s)
- K. E. Huus
- grid.17091.3e0000 0001 2288 9830Michael Smith Laboratories and Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC Canada
| | - T. T. Hoang
- grid.17091.3e0000 0001 2288 9830Michael Smith Laboratories and Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC Canada
| | - A. Creus-Cuadros
- grid.17091.3e0000 0001 2288 9830Michael Smith Laboratories and Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC Canada
| | - M. Cirstea
- grid.17091.3e0000 0001 2288 9830Michael Smith Laboratories and Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC Canada
| | - S. L. Vogt
- grid.17091.3e0000 0001 2288 9830Michael Smith Laboratories and Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC Canada
| | - K. Knuff-Janzen
- grid.17091.3e0000 0001 2288 9830Michael Smith Laboratories and Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC Canada
| | - P. J. Sansonetti
- grid.428999.70000 0001 2353 6535Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France ,grid.429007.80000 0004 0627 2381Present Address: Center for Microbes, Development and Health, Institut Pasteur de Shanghai, Shanghai, China
| | - P. Vonaesch
- grid.428999.70000 0001 2353 6535Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France ,grid.416786.a0000 0004 0587 0574Present Address: Human and Animal Health Unit, Swiss Tropical and Public Health Institute & University of Basel, Basel, Switzerland
| | - B. B. Finlay
- grid.17091.3e0000 0001 2288 9830Michael Smith Laboratories and Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC Canada ,grid.440050.50000 0004 0408 2525Canadian Institute for Advanced Research, Toronto, Ontario Canada
| |
Collapse
|
29
|
Sewunet T, Asrat D, Woldeamanuel Y, Ny S, Westerlund F, Aseffa A, Giske CG. Polyclonal spread of bla CTX-M-15 through high-risk clones of Escherichia coli at a tertiary hospital in Ethiopia. J Glob Antimicrob Resist 2021; 29:405-412. [PMID: 34775133 DOI: 10.1016/j.jgar.2021.09.017] [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] [Received: 09/14/2020] [Revised: 09/18/2021] [Accepted: 09/28/2021] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE The burden of antimicrobial resistance and spread of epidemic clones are rarely reported from low-income countries. We aimed to investigate genome-based epidemiology of extended-spectrum beta-lactamase-producing Escherichia coli (ESBL-EC) at a tertiary hospital in Jimma, Ethiopia. METHODS Bacteria were isolated from clinical specimens obtained from Jimma Medical Center and subjected to species identification (MALDI-TOF), antibiotic susceptibility testing (disk diffusion), and whole genome sequencing (Illumina HiSeq2500). Genomic data analysis was performed using the Enterobase and Center for Genomic Epidemiology bioinformatics pipelines. A maximum likelihood tree was generated using FastTree/2.1.8 based on SNPs in shared genomic regions to identify transmission clusters. RESULT E. coli isolates (n=261) were collected from 1,087 single non-repeat clinical specimens over a period of five months in 2016. The prevalence of ESBL-EC was (54.7%, 143/261), and 96% of these isolates were resistant to multiple classes of antibiotics. ESBL-gene blaCTX-M-15 was present in 88.4.% of the isolates (122/138). Genes conferring resistance to aminoglycosides and ciprofloxacin - aac(6')-Ib-cr (62.3%, 86/138), phenicols - catB3 (56.5%, 78/138), sulfonamides - sul1 (68.1%, 94/138), trimethoprim - dfrA17 (57.9%, 80/138) and macrolides - mph(A) (67.3%, 93/138) were detected. The most prevalent sequence types were ST410 (23%), ST648 (17%), ST131 (10%), and ST167 (7%). Isolates of same sequence type collected from different units of the hospital were highly similar in SNP-analysis. CONCLUSION A high prevalence of ESBL, and dissemination of blaCTX-M-15 through multiple high-risk clones of E. coli, was detected. The nosocomial spread of multidrug-resistant ESBL-EC within the hospital puts vulnerable patients at risk for difficult-to-treat infections.
Collapse
Affiliation(s)
- Tsegaye Sewunet
- Department of Microbiology, Immunology and Parasitology, Addis Ababa University, Ethiopia; School of Laboratory Sciences, Jimma University, Jimma, Ethiopia; Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, Sweden.
| | - Daniel Asrat
- Department of Microbiology, Immunology and Parasitology, Addis Ababa University, Ethiopia
| | | | - Sofia Ny
- Public Health Agency of Sweden, Sweden
| | - Fredrik Westerlund
- Department of Biology and Biological Engineering, Chalmers University of Technology, Sweden
| | - Abraham Aseffa
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Christian G Giske
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, Sweden; Karolinska University Hospital, Clinical Microbiology, Sweden
| |
Collapse
|
30
|
Keddy KH, Saha S, Okeke IN, Kalule JB, Qamar FN, Kariuki S. Combating Childhood Infections in LMICs: evaluating the contribution of Big Data Big data, biomarkers and proteomics: informing childhood diarrhoeal disease management in Low- and Middle-Income Countries. EBioMedicine 2021; 73:103668. [PMID: 34742129 PMCID: PMC8579132 DOI: 10.1016/j.ebiom.2021.103668] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 09/26/2021] [Accepted: 10/20/2021] [Indexed: 01/20/2023] Open
Abstract
Despite efforts to reduce the global burden of childhood diarrhoea, 50% of all cases globally occur in children under five years in Low–Income and Middle- Income Countries (LMICs) and knowledge gaps remain regarding the aetiological diagnosis, introduction of diarrhoeal vaccines, and the role of environmental enteric dysfunction and severe acute malnutrition. Biomarkers may assist in understanding disease processes, from diagnostics, to management of childhood diarrhoea and the sequelae to vaccine development. Proteomics has the potential to assist in the identification of new biomarkers to understand the processes in the development of childhood diarrhoea and to aid in developing new vaccines. Centralised repositories that enable mining of large data sets to better characterise risk factors, the proteome of both the patient and the different diarrhoeal pathogens, and the environment, could inform patient management and vaccine development, providing a systems biological approach to address the burden of childhood diarrhoea in LMICs.
Collapse
Affiliation(s)
- Karen H Keddy
- Tuberculosis Platform, South African Medical Research Council, 1 Soutpansberg Rd, Pretoria, 0001, South Africa.
| | - Senjuti Saha
- Child Health Research Foundation, 23/2 Khilji Road, Mohammadpur, Dhaka 1207, Bangladesh
| | - Iruka N Okeke
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Oyo State, Nigeria
| | - John Bosco Kalule
- Biotechnical and Diagnostic Sciences, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Uganda
| | - Farah Naz Qamar
- Department of Pediatrics and Child Health. Aga Khan University, Stadoum road Karachi, Pakistan 74800
| | - Samuel Kariuki
- Centre for Microbiology Research, Kenya Medical Research Institute, Off Mbagathi Road, Nairobi, Kenya
| |
Collapse
|
31
|
Chen D, Mechlowitz K, Li X, Schaefer N, Havelaar AH, McKune SL. Benefits and Risks of Smallholder Livestock Production on Child Nutrition in Low- and Middle-Income Countries. Front Nutr 2021; 8:751686. [PMID: 34778344 PMCID: PMC8579112 DOI: 10.3389/fnut.2021.751686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022] Open
Abstract
Livestock production may improve nutritional outcomes of pregnant women and children by increasing household income, availability of nutrient-dense foods, and women's empowerment. Nevertheless, the relationship is complex, and the nutritional status of children may be impaired by presence of or proximity to livestock and their pathogens. In this paper, we review the benefits and risks of livestock production on child nutrition. Evidence supports the nutritional benefits of livestock farming through income, production, and women's empowerment. Increasing animal source food consumption requires a combination of efforts, including improved animal management so that herd size is adequate to meet household income needs and consumption and addressing sociocultural and gendered norms. Evidence supports the inclusion of behavior change communication strategies into livestock production interventions to facilitate the sustainability of nutritional benefits over time, particularly interventions that engage women and foster dimensions of women's empowerment. In evaluating the risks of livestock production, evidence indicates that a broad range of enteric pathogens may chronically infect the intestines of children and, in combination with dietary deficits, may cause environmental enteric dysfunction (EED), a chronic inflammation of the gut. Some of the most important pathogens associated with EED are zoonotic in nature with livestock as their main reservoir. Very few studies have aimed to understand which livestock species contribute most to colonization with these pathogens, or how to reduce transmission. Control at the point of exposure has been investigated in a few studies, but much less effort has been spent on improving animal husbandry practices, which may have additional benefits. There is an urgent need for dedicated and long-term research to understand which livestock species contribute most to exposure of young children to zoonotic enteric pathogens, to test the potential of a wide range of intervention methods, to assess their effectiveness in randomized trials, and to assure their broad adaptation and sustainability. This review highlights the benefits and risks of livestock production on child nutrition. In addition to identifying research gaps, findings support inclusion of poor gut health as an immediate determinant of child undernutrition, expanding the established UNICEF framework which includes only inadequate diet and disease.
Collapse
Affiliation(s)
- Dehao Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Karah Mechlowitz
- Department of Social and Behavioral Sciences, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Xiaolong Li
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Nancy Schaefer
- Health Science Center Libraries, University of Florida, Gainesville, FL, United States
| | - Arie H. Havelaar
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Animal Sciences, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
- Institute for Sustainable Food Systems, University of Florida, Gainesville, FL, United States
| | - Sarah L. McKune
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
- Center for African Studies, University of Florida, Gainesville, FL, United States
| |
Collapse
|
32
|
Pakbin B, Brück WM, Rossen JWA. Virulence Factors of Enteric Pathogenic Escherichia coli: A Review. Int J Mol Sci 2021; 22:9922. [PMID: 34576083 PMCID: PMC8468683 DOI: 10.3390/ijms22189922] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 12/18/2022] Open
Abstract
Escherichia coli are remarkably versatile microorganisms and important members of the normal intestinal microbiota of humans and animals. This harmless commensal organism can acquire a mixture of comprehensive mobile genetic elements that contain genes encoding virulence factors, becoming an emerging human pathogen capable of causing a broad spectrum of intestinal and extraintestinal diseases. Nine definite enteric E. coli pathotypes have been well characterized, causing diseases ranging from various gastrointestinal disorders to urinary tract infections. These pathotypes employ many virulence factors and effectors subverting the functions of host cells to mediate their virulence and pathogenesis. This review summarizes new developments in our understanding of diverse virulence factors associated with encoding genes used by different pathotypes of enteric pathogenic E. coli to cause intestinal and extraintestinal diseases in humans.
Collapse
Affiliation(s)
- Babak Pakbin
- Institute for Life Technologies, University of Applied Sciences Western Switzerland Valais-Wallis, 1950 Sion 2, Switzerland;
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin 15315-3419, Iran
| | - Wolfram M. Brück
- Institute for Life Technologies, University of Applied Sciences Western Switzerland Valais-Wallis, 1950 Sion 2, Switzerland;
| | - John W. A. Rossen
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| |
Collapse
|
33
|
Soria-Bustos J, Saitz W, Medrano A, Lara-Ochoa C, Bennis Z, Monteiro-Neto V, Dos Santos CI, Rodrigues J, Hernandes RT, Yáñez JA, Torres J, Navarro-García F, Martínez-Laguna Y, Fontes Piazza RM, Munhoz DD, Cedillo ML, Ares MA, De la Cruz MA, Nataro JP, Girón JA. Role of the YehD fimbriae in the virulence-associated properties of enteroaggregative Escherichia coli. Environ Microbiol 2021; 24:1035-1051. [PMID: 34431194 DOI: 10.1111/1462-2920.15737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/16/2021] [Accepted: 08/22/2021] [Indexed: 11/30/2022]
Abstract
The interaction of enteroaggregative Escherichia coli (EAEC) strains with the colonic gut mucosa is characterized by the ability of the bacteria to form robust biofilms, to bind mucin, and induce a local inflammatory response. These events are mediated by a repertoire of five different aggregative adherence fimbriae variants (AAF/I-V) typically encoded on virulence plasmids. In this study, we report the production in EAEC strains of a new YehD fimbriae (YDF), which is encoded by the chromosomal gene cluster yehABCD, also present in most E. coli strains. Immuno-labelling of EAEC strain 042 with anti-AAF/II and anti-YDF antibodies demonstrated the presence of both AAF/II and YDF on the bacterial surface. We investigated the role of YDF in cell adherence, biofilm formation, colonization of spinach leaves, and induction of pro-inflammatory cytokines release. To this aim, we constructed yehD deletion mutants in different EAEC backgrounds (strains 17-2, 042, 55989, C1010, 278-1, J7) each harbouring one of the five AAFs. The effect of the YDF mutation was strain dependent and AAF independent as the lack of YDF had a different impact on the phenotypes manifested by the different EAECs tested. Expression of the yehABCD operon in a E. coli K12 ORN172 showed that YDF is important for biofilm formation but not for adherence to HeLa cells. Lastly, screening of pro-inflammatory cytokines in supernatants of Caco-2 cells infected with EAEC strains 042 and J7 and their isogenic ΔyehD mutants showed that these mutants were significantly defective in release of IL-8 and TNF-α. This study contributes to the understanding of the complex and diverse mechanisms of adherence of EAEC strains and identifies a new potential target for preventive measures of gastrointestinal illness caused by EAEC and other E. coli pathogroups.
Collapse
Affiliation(s)
- Jorge Soria-Bustos
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA.,Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Centro Médico Nacional Siglo XXI, Instituto Mexicano de Seguro Social, Ciudad de México, Mexico
| | - Waleska Saitz
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Abraham Medrano
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Cristina Lara-Ochoa
- Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Zineb Bennis
- University of Florida, Emerging Pathogens Institute, Gainesville, FL, USA
| | | | | | - Josias Rodrigues
- Departamento de Microbiologia e Imunologia, Instituto de Biociencias da UNESP, Botucatu, SP, Brazil
| | - Rodrigo T Hernandes
- Departamento de Microbiologia e Imunologia, Instituto de Biociencias da UNESP, Botucatu, SP, Brazil
| | - Jorge A Yáñez
- Facultad de Estomatología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Javier Torres
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Centro Médico Nacional Siglo XXI, Instituto Mexicano de Seguro Social, Ciudad de México, Mexico
| | - Fernando Navarro-García
- Departamento de Biología Celular, Centro de Investigaciones Avanzadas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Ygnacio Martínez-Laguna
- Centro de Investigaciones en Ciencias Microbiológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | | | | | - María L Cedillo
- Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Miguel A Ares
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Centro Médico Nacional Siglo XXI, Instituto Mexicano de Seguro Social, Ciudad de México, Mexico
| | - Miguel A De la Cruz
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Centro Médico Nacional Siglo XXI, Instituto Mexicano de Seguro Social, Ciudad de México, Mexico
| | - James P Nataro
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jorge A Girón
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA.,Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| |
Collapse
|
34
|
Vonaesch P, Djorie SG, Kandou KJE, Rakotondrainipiana M, Schaeffer L, Andriatsalama PV, Randriamparany R, Gondje BP, Nigatoloum S, Vondo SS, Etienne A, Robinson A, Hunald FA, Raharimalala L, Giles-Vernick T, Tondeur L, Randrianirina F, Bastaraud A, Gody JC, Sansonetti PJ, Randremanana RV. Factors Associated with Stunted Growth in Children Under Five Years in Antananarivo, Madagascar and Bangui, Central African Republic. Matern Child Health J 2021; 25:1626-1637. [PMID: 34383227 PMCID: PMC8448698 DOI: 10.1007/s10995-021-03201-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2021] [Indexed: 11/29/2022]
Abstract
Objectives With a fourth of all under-five children affected, stunting remains one of the biggest health challenges worldwide. Even though the main underlying factors are known, the exact pathways to stunting varying in affected regions, and interventions thus need to be tailored to the local contexts. This study aimed assessing and comparing factors associated with stunting in two understudied sub-Saharan urban contexts with some of the highest stunting prevalence globally: Bangui, Central African Republic (~ 36%) and Antananarivo, Madagascar (42%). Methods We performed a case–control study on 175 + 194 stunted and 237 + 230 non-stunted control children aged 2–5 years and matched for age, gender and district of residency. Factors associated with stunting were identified using a standardized, paper questionnaire delivered by trained interviewers. Statistical analysis was done using logistic regression modelling. Results In both sites, formal maternal education lowered the risk of being stunted and restricted access to soap, suffering of anaemia and low birth weight were associated with higher risk of stunting. Short maternal stature, household head different from parents, diarrhoea and coughing were associated with an increased risk and continuing breastfeeding was associated with a lower risk of stunting in Antananarivo. Previous severe undernutrition and dermatitis/ fungal skin infections were associated with higher and changes in diet during pregnancy with lower risk of stunting in Bangui. Conclusions Our results suggest maternal education, antenatal care, iron supplementation and simple WASH interventions such as using soap and infection control as general and breastfeeding (Antananarivo) or better nutrition (Bangui) as area-specified interventions. Supplementary Information The online version contains supplementary material available at 10.1007/s10995-021-03201-8.
Collapse
Affiliation(s)
- Pascale Vonaesch
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 25-28 Rue du Dr Roux, Paris, France. .,Human and Animal Health Unit, Swiss Tropical and Public Health Institute, Socinstrasse 57, 4051, Basel, Switzerland. .,University of Basel, 4051, Basel, Switzerland.
| | - Serge Ghislain Djorie
- Unité D'Epidémiologie, Institut Pasteur de Bangui, Avenue de l'Indépendance, Bangui, Central African Republic
| | - Kaleb Jephté Estimé Kandou
- Unité D'Epidémiologie, Institut Pasteur de Bangui, Avenue de l'Indépendance, Bangui, Central African Republic
| | - Maheninasy Rakotondrainipiana
- Unité Epidémiologie Et de Recherche Clinique, Institut Pasteur de Madagascar-Ambatofotsikely, BP 1274, 101, Antananarivo, Madagascar
| | - Laura Schaeffer
- Unité D'Epidémiologie Des Maladies Emergentes, Institut Pasteur, 28 Rue du Dr. Roux, 75015, Paris, France
| | - Prisca Vega Andriatsalama
- Unité Epidémiologie Et de Recherche Clinique, Institut Pasteur de Madagascar-Ambatofotsikely, BP 1274, 101, Antananarivo, Madagascar
| | - Ravaka Randriamparany
- Unité Epidémiologie Et de Recherche Clinique, Institut Pasteur de Madagascar-Ambatofotsikely, BP 1274, 101, Antananarivo, Madagascar
| | - Bolmbaye Privat Gondje
- Centre Pédiatrique de Bangui, Avenue de l'Indépendance, Bangui, Central African Republic
| | - Synthia Nigatoloum
- Centre Pédiatrique de Bangui, Avenue de l'Indépendance, Bangui, Central African Republic
| | - Sonia Sandrine Vondo
- Centre Pédiatrique de Bangui, Avenue de l'Indépendance, Bangui, Central African Republic
| | - Aurélie Etienne
- Unité Epidémiologie Et de Recherche Clinique, Institut Pasteur de Madagascar-Ambatofotsikely, BP 1274, 101, Antananarivo, Madagascar
| | - Annick Robinson
- Centre Hospitalier Universitaire Mère Enfant de Tsaralalana, Rue Patrice Lumumba, Rue Mabizo S, 101, Antananarivo, Madagascar
| | - Francis Allen Hunald
- Service de Chirurgie Pédiatrique, Centre Hospitalier Universitaire Joseph Ravoahangy Andrianavalona, Ampefiloha, BP 4150, 101, Antananarivo, Madagascar
| | - Lisette Raharimalala
- Centre de Santé Maternelle Et Infantile de Tsaralalana, Lalana Andriantsilavo, 101, Antananarivo, Madagascar
| | - Tamara Giles-Vernick
- Anthropology and Ecology of Disease Emergence Unit, Institut Pasteur, 28 Rue du Dr. Roux, 75015, Paris, France
| | - Laura Tondeur
- Unité D'Epidémiologie Des Maladies Emergentes, Institut Pasteur, 28 Rue du Dr. Roux, 75015, Paris, France
| | - Frédérique Randrianirina
- Centre de Biologie Clinique, Institut Pasteur de Madagascar, BP 1274, 101, Antananarivo, Madagascar
| | - Alexandra Bastaraud
- Laboratoire D'Hygiène Des Aliments Et de L'Environnement (LHAE), Institut Pasteur de Madagascar, BP 1274, 101, Antananarivo, Madagascar
| | - Jean-Chrysostome Gody
- Centre Pédiatrique de Bangui, Avenue de l'Indépendance, Bangui, Central African Republic
| | - Philippe Jean Sansonetti
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 25-28 Rue du Dr Roux, Paris, France.,The Center for Microbes, Development and Health, Institut Pasteur of Shanghai and Chinese Academy of Sciences, 411 Hefei Rd, Huangpu, Shanghai, China
| | - Rindra Vatosoa Randremanana
- Unité Epidémiologie Et de Recherche Clinique, Institut Pasteur de Madagascar-Ambatofotsikely, BP 1274, 101, Antananarivo, Madagascar.
| | | |
Collapse
|
35
|
Vergis J, Malik SVS, Pathak R, Kumar M, Sunitha R, Barbuddhe SB, Rawool DB. Efficacy of Indolicidin, Cecropin A (1-7)-Melittin (CAMA) and Their Combination Against Biofilm-Forming Multidrug-Resistant Enteroaggregative Escherichia coli. Probiotics Antimicrob Proteins 2021; 12:705-715. [PMID: 31485973 DOI: 10.1007/s12602-019-09589-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The present study examined the anti-biofilm efficacy of two short-chain antimicrobial peptides (AMPs), namely, indolicidin and cecropin A (1-7)-melittin (CAMA) against biofilm-forming multidrug-resistant enteroaggregative Escherichia coli (MDR-EAEC) isolates. The typical EAEC isolates re-validated by PCR and confirmed using HEp-2 cell adherence assay was subjected to antibiotic susceptibility testing to confirm its MDR status. The biofilm-forming ability of MDR-EAEC isolates was assessed by Congo red binding, microtitre plate assays and hydrophobicity index; broth microdilution technique was employed to determine minimum inhibitory concentrations (MICs) and minimum biofilm eradication concentrations (MBECs). The obtained MIC and MBEC values for both AMPs were evaluated alone and in combination against MDR-EAEC biofilms using crystal violet (CV) staining and confocal microscopy-based live/dead cell quantification methods. All the three MDR-EAEC strains revealed weak to strong biofilm-forming ability and were found to be electron-donating and weakly electron-accepting (hydrophobicity index). Also, highly significant (P < 0.001) time-dependent hydrodynamic growth of the three MDR-EAEC strains was observed at 48 h of incubation in Dulbecco's modified Eagle's medium (DMEM) containing 0.45% D-glucose. AMPs and their combination were able to inhibit the initial biofilm formation at 24 h and 48 h as evidenced by CV staining and confocal quantification. Further, the application of AMPs (individually and combination) against the preformed MDR-EAEC biofilms resulted in highly significant eradication (P < 0.001) at 24 h post treatment. However, significant differences were not observed between AMP treatments (individually or in combination). The AMPs seem to be an effective candidates for further investigations such as safety, stability and appropriate biofilm-forming MDR-EAEC animal models.
Collapse
Affiliation(s)
- Jess Vergis
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - S V S Malik
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Richa Pathak
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Manesh Kumar
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - R Sunitha
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - S B Barbuddhe
- ICAR-National Research Centre on Meat, Chengicherla, Telangana, 500092, India
| | - Deepak B Rawool
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India.
| |
Collapse
|
36
|
Modgil V, Chaudhary P, Bharti B, Mahindroo J, Yousuf M, Koundal M, Mohan B, Taneja N. Prevalence, Virulence Gene Profiling, and Characterization of Enteroaggregative Escherichia coli from Children with Acute Diarrhea, Asymptomatic Nourished, and Malnourished Children Younger Than 5 Years of Age in India. J Pediatr 2021; 234:106-114.e5. [PMID: 33713662 DOI: 10.1016/j.jpeds.2021.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To study the significance of enteroaggregative Escherichia coli (EAEC) as a pathogen causing acute diarrhea and a commensal in healthy nourished and malnourished children younger than five years of age in the Chandigarh region and to address possible traits of EAEC virulence genes, biofilm formation, phylogroups, and antibiotic resistance that would be correlated with diarrhea or carriage. STUDY DESIGN Stool samples were obtained from children with acute diarrhea (n = 548), as well as nourished (n = 550), and malnourished controls without diarrhea (n = 110). E coli isolates were confirmed as EAEC by pCVD432 polymerase chain reaction. Multiplex polymerase chain reactions were used to identify 22 virulence-related genes and phylogeny. Antibiotic susceptibility, adherence, and biofilm-forming potential also were studied. RESULTS Overall, 16.6% of children were malnourished. EAEC detection was greater among children with acute diarrhea (16%) than nourished (6%) and malnourished nondiarrheal controls (2.7%). We found an association of EAEC infections with age <2 years (P = .0001) in the diarrheal group. Adhesive variants adhesion fimbriae IV and adhesion fimbriae II were significantly associated with diarrhea. The aggR and aar genes showed a positive and negative association with the severity of disease (P = .0004 and P = .0003). A high degree of multidrug resistance was found (73.8%) in the diarrheal group. Most EAEC strains from the diarrheal group belonged to B2 and D phylogroups, whereas strains from non-diarrheal groups, which belonged to phylogroup B1. CONCLUSIONS EAEC is a significant contributor to childhood diarrhea, its presence as a commensal, and the significance of the association of various virulence factors among the EAEC isolated from diarrheal and non-diarrheal stools. These data reinforce the importance of aggR and aar as positive and negative regulators and the contribution of AAF/II and AAF/IV fimbria for the pathobiology of EAEC.
Collapse
Affiliation(s)
- Vinay Modgil
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Chaudhary
- Department of Pediatrics Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bhavneet Bharti
- Department of Pediatrics Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jaspreet Mahindroo
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Md Yousuf
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Meenakshi Koundal
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Balvinder Mohan
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Neelam Taneja
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Several types of Escherichia coli cause acute diarrhea in humans and are responsible for a large burden of disease globally. The purpose of this review is to summarize diarrheagenic Escherichia coli (DEC) pathotype definitions and discuss existing and emerging molecular, genomic, and gut microbiome methods to detect, define, and study DEC pathotypes. RECENT FINDINGS DEC pathotypes are currently diagnosed by molecular detection of unique virulence genes. However, some pathotypes have defied coherent molecular definitions because of imperfect gene targets, and pathotype categories are complicated by hybrid strains and isolation of pathotypes from asymptomatic individuals. Recent progress toward more efficient, sensitive, and multiplex DEC pathotype detection has been made using emerging PCR-based technologies. Genomics and gut microbiome detection methods continue to advance rapidly and are contributing to a better understanding of DEC pathotype diversity and functional potential. SUMMARY DEC pathotype categorizations and detection methods are useful but imperfect. The implementation of molecular and sequence-based methods and well designed epidemiological studies will continue to advance understanding of DEC pathotypes. Additional emphasis is needed on sequencing DEC genomes from regions of the world where they cause the most disease and from the pathotypes that cause the greatest burden of disease globally.
Collapse
|
38
|
Guerrant RL, Bolick DT, Swann JR. Modeling Enteropathy or Diarrhea with the Top Bacterial and Protozoal Pathogens: Differential Determinants of Outcomes. ACS Infect Dis 2021; 7:1020-1031. [PMID: 33901398 PMCID: PMC8154416 DOI: 10.1021/acsinfecdis.0c00831] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Developing effective
therapeutics or preventive interventions for
important health threats is greatly enhanced whenever accessible models
can enable the assessment of clinically important outcomes. While
no non-human model is ever perfect, inexpensive in vivo small animal models in such as mice are often of great help in assessing
the relevant efficacy of potential interventions. In addition to acute
diarrhea, the long-term growth and developmental effects of enteric
infections, with or without overt diarrhea, are increasingly recognized.
To address these diverse effects, inexpensive animal models are proving
to be very helpful. Herein, we review the major clinical concerns
with enteric parasitic and bacterial infections that are extremely
common worldwide, especially in vulnerable young children living in
impoverished areas, and the recently published murine models of these
infections and their outcomes. We find that common dietary deficiencies
seen in children in developing areas have striking effects on diarrhea
and enteropathy outcomes in mice. However, these effects differ with
different pathogens. Specifically, the effects of protein or zinc
deficiency differ considerably with different major protozoal and
bacterial pathogens, suggesting different pathogenetic pathways and
intervention effects. The pathogens reviewed are the seven top parasitic
and bacterial pathogens seen in children, namely, Cryptosporidium, Giardia, Campylobacter, Shigella, enterotoxigenic Escherichia coli (ETEC), enteroaggregative E. coli (EAEC), and enteropathogenic E. coli (EPEC).
Collapse
Affiliation(s)
- Richard L. Guerrant
- Center for Global Health Equity, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - David T. Bolick
- Center for Global Health Equity, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - Jonathan R. Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
- Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
39
|
Patterson GT, Manthi D, Osuna F, Muia A, Olack B, Mbuchi M, Saldarriaga OA, Ouma L, Inziani M, Yu X, Otieno P, Melby PC. Environmental, Metabolic, and Inflammatory Factors Converge in the Pathogenesis of Moderate Acute Malnutrition in Children: An Observational Cohort Study. Am J Trop Med Hyg 2021; 104:1877-1888. [PMID: 33755580 PMCID: PMC8103470 DOI: 10.4269/ajtmh.20-0963] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/07/2021] [Indexed: 01/04/2023] Open
Abstract
Acute malnutrition affects more than 50 million children worldwide. These children are at an increased risk of morbidity and mortality from infectious disease. However, the pathogenesis of acute malnutrition and mechanisms underlying the increased risk and poor outcomes from infection are not well understood. Our objective was to identify differences in inflammation and inflammatory responses between children with moderate acute malnutrition (MAM) and healthy controls (HCs), and search for environmental, pathophysiological, and metabolic factors that may influence this response. Sixteen children with MAM and 16 HCs aged 18-36 months were studied in Nairobi, Kenya. None of the children had symptoms of an infectious disease (fever, diarrhea, or cough) in the 2 weeks before enrollment and sample collection. Demographic and health data were provided by their primary caregivers. Blood samples were collected to measure various biomarkers and the response to an inflammatory stimulus. Children with MAM were more frequently from households with contaminated water, crowding, and unstable income sources. They also had increases in basal inflammation, circulating bacterial lipopolysaccharide (LPS), markers of intestinal damage, and an exaggerated whole blood inflammatory response to LPS. Metabolic changes in children with MAM led to increased plasma levels of long-chain fatty acids, which were found to contribute to the pro-inflammatory state. These exploratory findings suggest convergence of multiple factors to promote dysregulated inflammatory responses and prompt several mechanistic hypotheses that can be pursued to better understand the pathogenesis of MAM.
Collapse
Affiliation(s)
- Grace T. Patterson
- Department of Internal Medicine and Infectious Disease, University of Texas Medical Branch, Galveston, Texas
| | - Dennis Manthi
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Finnley Osuna
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Alfred Muia
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Beatrice Olack
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Margaret Mbuchi
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Omar A. Saldarriaga
- Department of Internal Medicine and Infectious Disease, University of Texas Medical Branch, Galveston, Texas
| | - Linet Ouma
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Mary Inziani
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Xiaoying Yu
- Department of Preventative Medicine and Population Health, University of Texas Medical Branch, Galveston, Texas
| | - Phelgona Otieno
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya;,Address correspondence to Phelgona Otieno, Kenya Medical Research Institute, Mbagathi Road, Nairobi, Kenya, E-mail: or Peter C. Melby, Department of Internal Medicine and Infectious Disease, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, E-mail:
| | - Peter C. Melby
- Department of Internal Medicine and Infectious Disease, University of Texas Medical Branch, Galveston, Texas;,Address correspondence to Phelgona Otieno, Kenya Medical Research Institute, Mbagathi Road, Nairobi, Kenya, E-mail: or Peter C. Melby, Department of Internal Medicine and Infectious Disease, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, E-mail:
| |
Collapse
|
40
|
Rogawski McQuade ET, Liu J, Kang G, Kosek MN, Lima AAM, Bessong PO, Samie A, Haque R, Mduma ER, Shrestha S, Leite JP, Bodhidatta L, Iqbal N, Page N, Kiwelu I, Bhutta Z, Ahmed T, Houpt ER, Platts-Mills JA. Protection From Natural Immunity Against Enteric Infections and Etiology-Specific Diarrhea in a Longitudinal Birth Cohort. J Infect Dis 2021; 222:1858-1868. [PMID: 31984416 PMCID: PMC7653087 DOI: 10.1093/infdis/jiaa031] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/22/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The degree of protection conferred by natural immunity is unknown for many enteropathogens, but it is important to support the development of enteric vaccines. METHODS We used the Andersen-Gill extension of the Cox model to estimate the effects of previous infections on the incidence of subsequent subclinical infections and diarrhea in children under 2 using quantitative molecular diagnostics in the MAL-ED cohort. We used cross-pathogen negative control associations to correct bias due to confounding by unmeasured heterogeneity of exposure and susceptibility. RESULTS Prior rotavirus infection was associated with a 50% lower hazard (calibrated hazard ratio [cHR], 0.50; 95% confidence interval [CI], 0.41-0.62) of subsequent rotavirus diarrhea. Strong protection was evident against Cryptosporidium diarrhea (cHR, 0.32; 95% CI, 0.20-0.51). There was also protection due to prior infections for norovirus GII (cHR against diarrhea, 0.67; 95% CI, 0.49-0.91), astrovirus (cHR, 0.62; 95% CI, 0.48-0.81), and Shigella (cHR, 0.79; 95% CI, 0.65-0.95). Minimal protection was observed for other bacteria, adenovirus 40/41, and sapovirus. CONCLUSIONS Natural immunity was generally stronger for the enteric viruses than bacteria, potentially due to less antigenic diversity. Vaccines against major causes of diarrhea may be feasible but likely need to be more immunogenic than natural infection.
Collapse
Affiliation(s)
- Elizabeth T Rogawski McQuade
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virgina, USA.,Division of Infectious Diseases & International Health, University of Virginia, Charlottesville, Virgina, USA
| | - Jie Liu
- Division of Infectious Diseases & International Health, University of Virginia, Charlottesville, Virgina, USA
| | | | - Margaret N Kosek
- Division of Infectious Diseases & International Health, University of Virginia, Charlottesville, Virgina, USA.,Asociación Benéfica PRISMA, Iquitos, Peru
| | | | | | | | - Rashidul Haque
- International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | | | | | | | - Ladaporn Bodhidatta
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | | | - Nicola Page
- National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Ireen Kiwelu
- Kilimanjaro Clinical Research Institute, Moshi, Tanzania
| | | | - Tahmeed Ahmed
- International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - Eric R Houpt
- Division of Infectious Diseases & International Health, University of Virginia, Charlottesville, Virgina, USA
| | - James A Platts-Mills
- Division of Infectious Diseases & International Health, University of Virginia, Charlottesville, Virgina, USA
| |
Collapse
|
41
|
The contribution of environmental enteropathy to the global problem of micronutrient deficiency. Proc Nutr Soc 2021; 80:303-310. [PMID: 33663621 DOI: 10.1017/s0029665121000549] [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: 01/20/2023]
Abstract
Sometimes referred to as hidden hunger, micronutrient deficiencies persist on a global scale. For some micronutrients this appears to be due to inadequate intake, for others intake may not match increased requirements. However, for most micronutrient deficiencies there is uncertainty as to the dominant driver, and the question about the contribution of malabsorption is open. Environmental enteropathy (EE), formerly referred to as tropical enteropathy and also referred to as environmental enteric dysfunction, is an asymptomatic disorder of small intestinal structure and function which is very highly prevalent in many disadvantaged populations. Recent studies of the pathology and microbiology of this disorder suggest that it is driven by very high pathogen burdens in children and adults living in insanitary environments and is characterised by major derangements of the epithelial cells of the intestinal mucosa. Transcriptomic data suggest that it may lead to impaired digestion and absorption of macronutrients. Given the very high prevalence of EE, marginal malabsorption could have large impacts at population scales. However, the relative contributions of inadequate soil and crop micronutrient contents, inadequate intake, malabsorption and increased requirements are unknown. Malabsorption may compromise attempts to improve micronutrient status, but with the exception of zinc there is currently little evidence to confirm that malabsorption contributes to micronutrient deficiency. Much further research is required to understand the role of malabsorption in hidden hunger, especially in very disadvantaged populations where these deficiencies are most prevalent.
Collapse
|
42
|
Molecular Epidemiology of Enteroaggregative Escherichia coli (EAEC) Isolates of Hospitalized Children from Bolivia Reveal High Heterogeneity and Multidrug-Resistance. Int J Mol Sci 2020; 21:ijms21249543. [PMID: 33334000 PMCID: PMC7765457 DOI: 10.3390/ijms21249543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/10/2020] [Accepted: 12/12/2020] [Indexed: 12/16/2022] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) is an emerging pathogen frequently associated with acute diarrhea in children and travelers to endemic regions. EAEC was found the most prevalent bacterial diarrheal pathogen from hospitalized Bolivian children less than five years of age with acute diarrhea from 2007 to 2010. Here, we further characterized the epidemiology of EAEC infection, virulence genes, and antimicrobial susceptibility of EAEC isolated from 414 diarrheal and 74 non-diarrheal cases. EAEC isolates were collected and subjected to a PCR-based virulence gene screening of seven virulence genes and a phenotypic resistance test to nine different antimicrobials. Our results showed that atypical EAEC (a-EAEC, AggR-negative) was significantly associated with diarrhea (OR, 1.62, 95% CI, 1.25 to 2.09, p < 0.001) in contrast to typical EAEC (t-EAEC, AggR-positive). EAEC infection was most prevalent among children between 7–12 months of age. The number of cases exhibited a biannual cycle with a major peak during the transition from warm to cold (April–June). Both typical and a-EAEC infections were graded as equally severe; however, t-EAEC harbored more virulence genes. aap, irp2 and pic were the most prevalent genes. Surprisingly, we detected 60% and 52.6% of multidrug resistance (MDR) EAEC among diarrheal and non-diarrheal cases. Resistance to ampicillin, sulfonamides, and tetracyclines was most common, being the corresponding antibiotics, the ones that are frequently used in Bolivia. Our work is the first study that provides comprehensive information on the high heterogenicity of virulence genes in t-EAEC and a- EAEC and the large prevalence of MDR EAEC in Bolivia.
Collapse
|
43
|
Modgil V, Mahindroo J, Narayan C, Kalia M, Yousuf M, Shahi V, Koundal M, Chaudhary P, Jain R, Sandha KS, Tanwar S, Gupta P, Thakur K, Singh D, Gautam N, Kakkar M, Bharti B, Mohan B, Taneja N. Comparative analysis of virulence determinants, phylogroups, and antibiotic susceptibility patterns of typical versus atypical Enteroaggregative E. coli in India. PLoS Negl Trop Dis 2020; 14:e0008769. [PMID: 33206643 PMCID: PMC7673547 DOI: 10.1371/journal.pntd.0008769] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 09/01/2020] [Indexed: 11/30/2022] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) is an evolving enteric pathogen that causes acute and chronic diarrhea in developed and industrialized nations in children. EAEC epidemiology and the importance of atypical EAEC (aEAEC) isolation in childhood diarrhea are not well documented in the Indian setting. A comparative analysis was undertaken to evaluate virulence, phylogeny, and antibiotic sensitivity among typical tEAEC versus aEAEC. A total of 171 EAEC isolates were extracted from a broad surveillance sample of diarrheal (N = 1210) and healthy children (N = 550) across North India. Polymerase chain reaction (PCR) for the aggR gene (master regulator gene) was conducted to differentiate tEAEC and aEAEC. For 21 virulence genes, we used multiplex PCR to classify possible virulence factors among these strains. Phylogenetic classes were identified by a multiplex PCR for chuA, yjaA, and a cryptic DNA fragment, TspE4C2. Antibiotic susceptibility was conducted by the disc diffusion method as per CLSI guidelines. EAEC was associated with moderate to severe diarrhea in children. The prevalence of EAEC infection (11.4%) was higher than any other DEC group (p = 0.002). tEAEC occurrence in the diarrheal group was higher than in the control group (p = 0.0001). tEAEC strain harbored more virulence genes than aEAEC. astA, aap, and aggR genes were most frequently found in the EAEC from the diarrheal population. Within tEAEC, this gene combination was present in more than 50% of strains. Also, 75.8% of EAEC strains were multidrug-resistant (MDR). Phylogroup D (43.9%) and B1 (39.4%) were most prevalent in the diarrheal and control group, respectively. Genetic analysis revealed EAEC variability; the comparison of tEAEC and aEAEC allowed us to better understand the EAEC virulence repertoire. Further microbiological and epidemiological research is required to examine the pathogenicity of not only typical but also atypical EAEC. Enteroaggregative E. coli (EAEC) are an increasingly important cause of diarrhea. E. coli belonging to this category cause watery diarrhea, which is often persistent and can be inflammatory. It is also associated with traveler’s diarrhea in children and adults in middle and high-income countries. EAEC are defined by their ability to adhere to epithelial cells in a characteristic stacked brick-like pattern. However, the identification of these pathogenic strains remains elusive because of its heterogeneous nature. Genes that could contribute to the pathogenicity of EAEC encode adhesions, toxins, and other factors. Due to the heterogeneity of EAEC strains and differing host immune responses, not all EAEC infections are symptomatic. A critical factor in both recognizing EAEC pathogenesis and defining typical EAEC (tEAEC) strains is AggR, a transcriptional control for many EAEC virulence genes. The central role of aggR in virulence confers a strong priority to understand its pathogenicity. To identify EAEC, the CVD432 probe has been used. The CVD432 is a DNA probe from pAA plasmid of EAEC, has been reported to be specific for the detection of EAEC. The lack of sensitivity comes from the genetic heterogeneity of the EAEC strains and the wide geographic dispersal of strains. In our study, we performed a large surveillance of EAEC from North India among the pediatric population. Samples were collected by the microbiology staff at the Postgraduate Institute of Medical Education and Research (PGIMER) and referral system labs in Chandigarh (Manimajra), Punjab (Ludhiana), Haryana (Panchkula and Ambala Cantt), Himachal Pradesh (Hamirpur, Shimla, and Tanda), and Uttarakhand (Rishikesh, Rudrapur, and Haridwar)]. PGIMER is the largest tertiary care hospital in North India and serves patients from across Punjab, Jammu and Kashmir, Himachal Pradesh and Haryana. EAEC infections were detected using molecular methods. In our finding, astA, aap, and aggR genes were most frequently found in the EAEC from the diarrheal population. Within tEAEC, this gene combination is present in more than 50% of strains and helps to differentiate tEAEC from aEAEC. Our collection of EAEC strains helps in finding an appropriate marker for the early detection of EAEC. Our signature sequence (astA, aap, and aggR) will be ideal as focus genes for EAEC identification, as well as tEAEC and aEAEC. The multidrug resistance (MDR) was observed in 75.8% of the EAEC strains. tEAEC exhibits resistance to a greater number of antibiotics with respect to aEAEC. The phylogenetic analysis revealed that EAEC phylogeny is diverse and dispersed in all the phylogroups.
Collapse
Affiliation(s)
- Vinay Modgil
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Jaspreet Mahindroo
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Chandradeo Narayan
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Manmohit Kalia
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Md Yousuf
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Varun Shahi
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Meenakshi Koundal
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Pankaj Chaudhary
- Department of Pediatrics Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh (PGIMER), India
| | - Ruby Jain
- Civil Hospital Manimajra, Chandigarh, India
| | | | | | - Pratima Gupta
- Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Rishikesh, Uttarakhand, India
| | - Kamlesh Thakur
- Department of Microbiology, Dr. Rajendra Prasad Government Medical College Kangra (RPGMC), Himachal Pradesh, India
| | - Digvijay Singh
- Department of Microbiology, Indira Gandhi Medical college (IGMC), Shimla, Himachal Pradesh, India
| | - Neha Gautam
- Department of Microbiology, Indira Gandhi Medical college (IGMC), Shimla, Himachal Pradesh, India
| | | | - Bhavneet Bharti
- Department of Pediatrics Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh (PGIMER), India
| | - Balvinder Mohan
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Neelam Taneja
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
- * E-mail:
| |
Collapse
|
44
|
Boisen N, Østerlund MT, Joensen KG, Santiago AE, Mandomando I, Cravioto A, Chattaway MA, Gonyar LA, Overballe-Petersen S, Stine OC, Rasko DA, Scheutz F, Nataro JP. Redefining enteroaggregative Escherichia coli (EAEC): Genomic characterization of epidemiological EAEC strains. PLoS Negl Trop Dis 2020; 14:e0008613. [PMID: 32898134 PMCID: PMC7500659 DOI: 10.1371/journal.pntd.0008613] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 09/18/2020] [Accepted: 07/20/2020] [Indexed: 11/19/2022] Open
Abstract
Although enteroaggregative E. coli (EAEC) has been implicated as a common cause of diarrhea in multiple settings, neither its essential genomic nature nor its role as an enteric pathogen are fully understood. The current definition of this pathotype requires demonstration of cellular adherence; a working molecular definition encompasses E. coli which do not harbor the heat-stable or heat-labile toxins of enterotoxigenic E. coli (ETEC) and harbor the genes aaiC, aggR, and/or aatA. In an effort to improve the definition of this pathotype, we report the most definitive characterization of the pan-genome of EAEC to date, applying comparative genomics and functional characterization on a collection of 97 EAEC strains isolated in the course of a multicenter case-control diarrhea study (Global Enteric Multi-Center Study, GEMS). Genomic analysis revealed that the EAEC strains mapped to all phylogenomic groups of E. coli. Circa 70% of strains harbored one of the five described AAF variants; there were no additional AAF variants identified, and strains that lacked an identifiable AAF generally did not have an otherwise complete AggR regulon. An exception was strains that harbored an ETEC colonization factor (CF) CS22, like AAF a member of the chaperone-usher family of adhesins, but not phylogenetically related to the AAF family. Of all genes scored, sepA yielded the strongest association with diarrhea (P = 0.002) followed by the increased serum survival gene, iss (p = 0.026), and the outer membrane protease gene ompT (p = 0.046). Notably, the EAEC genomes harbored several genes characteristically associated with other E. coli pathotypes. Our data suggest that a molecular definition of EAEC could comprise E. coli strains harboring AggR and a complete AAF(I-V) or CS22 gene cluster. Further, it is possible that strains meeting this definition could be both enteric bacteria and urinary/systemic pathogens.
Collapse
Affiliation(s)
- Nadia Boisen
- Statens Serum Institut, Department of Bacteria, Parasites and Fungi, Copenhagen, Denmark
| | - Mark T. Østerlund
- Statens Serum Institut, Department of Bacteria, Parasites and Fungi, Copenhagen, Denmark
| | - Katrine G. Joensen
- Statens Serum Institut, Department of Bacteria, Parasites and Fungi, Copenhagen, Denmark
| | - Araceli E. Santiago
- University of Virginia School of Medicine, Department of Pediatrics, Charlottesville, Virginia, United States of America
| | - Inacio Mandomando
- Centro de Investigação em Saúde da Manhiça (CISM), Maputo, Mozambique
| | - Alejandro Cravioto
- Universidad Nacional Autónoma de México, Faculty of Medicine, Mexico City, Mexico
| | - Marie A. Chattaway
- Public Health England, Gastrointestinal Bacteria Reference Unit (GBRU), Colindale, United Kingdom
| | - Laura A. Gonyar
- University of Virginia School of Medicine, Department of Pediatrics, Charlottesville, Virginia, United States of America
| | | | - O. Colin Stine
- University of Maryland School of Medicine, Department of Epidemiology and Public Health, Baltimore, Maryland, United States of America
| | - David A. Rasko
- University of Maryland School of Medicine, Institute for Genome Sciences, Department of Microbiology and Immunology, Baltimore, Maryland, United States of America
| | - Flemming Scheutz
- Statens Serum Institut, Department of Bacteria, Parasites and Fungi, Copenhagen, Denmark
| | - James P. Nataro
- University of Virginia School of Medicine, Department of Pediatrics, Charlottesville, Virginia, United States of America
| |
Collapse
|
45
|
Aggregative Adherence Fimbriae II of Enteroaggregative Escherichia coli Are Required for Adherence and Barrier Disruption during Infection of Human Colonoids. Infect Immun 2020; 88:IAI.00176-20. [PMID: 32631917 DOI: 10.1128/iai.00176-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/30/2020] [Indexed: 01/07/2023] Open
Abstract
Symptomatic and asymptomatic infection with the diarrheal pathogen enteroaggregative Escherichia coli (EAEC) is associated with growth faltering in children in developing settings. The mechanism of this association is unknown, emphasizing a need for better understanding of the interactions between EAEC and the human gastrointestinal mucosa. In this study, we investigated the role of the aggregative adherence fimbriae II (AAF/II) in EAEC adherence and pathogenesis using human colonoids and duodenal enteroids. We found that a null mutant in aafA, the major subunit of AAF/II, adhered significantly less than wild-type (WT) EAEC strain 042, and adherence was restored in a complemented strain. Immunofluorescence confocal microscopy of differentiated colonoids, which produce an intact mucus layer comprised of the secreted mucin MUC2, revealed bacteria at the epithelial surface and within the MUC2 layer. The WT strain adhered to the epithelial surface, whereas the aafA deletion strain remained within the MUC2 layer, suggesting that the presence or absence of AAF/II determines both the abundance and location of EAEC adherence. In order to determine the consequences of EAEC adherence on epithelial barrier integrity, colonoid monolayers were exposed to EAEC constructs expressing or lacking aafA Colonoids infected with WT EAEC had significantly decreased epithelial resistance, an effect that required AAF/II, suggesting that binding of EAEC to the epithelium is necessary to impair barrier function. In summary, we show that production of AAF/II is critical for adherence and barrier disruption in human colonoids, suggesting a role for this virulence factor in EAEC colonization of the gastrointestinal mucosa.
Collapse
|
46
|
Beczkiewicz A, Cebelinski E, Decuir M, Lappi V, Wang X, Smith K, Boxrud D, Medus C. High Relative Frequency of Enteroaggregative Escherichia coli Among Patients With Reportable Enteric Pathogens, Minnesota, 2016-2017. Clin Infect Dis 2020; 69:473-479. [PMID: 30321302 DOI: 10.1093/cid/ciy890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/10/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Enteroaggregative Escherichia coli (EAEC) is increasingly recognized as an enteric pathogen as clinical laboratories transition to culture-independent diagnostic tests that detect EAEC. To date, epidemiological studies have focused on children aged <5 years, and information on EAEC incidence, illness outcomes, and transmission avenues is limited. METHODS Enteric disease surveillance data in Minnesota were used to describe EAEC illnesses reported to the Minnesota Department of Health from September 2016 through August 2017. We determined laboratory characteristics of EAEC using pulsed-field gel electrophoresis and next-generation sequencing. Frequency of EAEC illness, demographic profile of cases, clinical characteristics of illness, and plausible food or environmental exposures leading to EAEC transmission were assessed. RESULTS During the study period, 329 EAEC cases were reported. Among a subset of health systems able to detect EAEC over the entire study, EAEC was the second most common reportable enteric pathogen detected after Campylobacter and the most detected diarrheagenic E. coli pathotype. No other reportable enteric pathogens were detected among 75.3% of EAEC cases, and 68% of cases reported no international travel before onset. Several virulence genes were associated with clinical characteristics. CONCLUSIONS We provide evidence that EAEC is a likely causative agent of diarrheal illness in the United States. Our study contributes to criteria development for identification of pathogenic EAEC and proposes potential exposure avenues.
Collapse
Affiliation(s)
- Aaron Beczkiewicz
- Foodborne, Waterborne, Vectorborne, and Zoonotic Diseases Section.,Public Health Laboratory, Minnesota Department of Health, St. Paul
| | | | - Marijke Decuir
- Foodborne, Waterborne, Vectorborne, and Zoonotic Diseases Section
| | - Victoria Lappi
- Public Health Laboratory, Minnesota Department of Health, St. Paul
| | - Xiong Wang
- Public Health Laboratory, Minnesota Department of Health, St. Paul
| | - Kirk Smith
- Foodborne, Waterborne, Vectorborne, and Zoonotic Diseases Section
| | - Dave Boxrud
- Public Health Laboratory, Minnesota Department of Health, St. Paul
| | - Carlota Medus
- Foodborne, Waterborne, Vectorborne, and Zoonotic Diseases Section
| |
Collapse
|
47
|
Mucus layer modeling of human colonoids during infection with enteroaggragative E. coli. Sci Rep 2020; 10:10533. [PMID: 32601325 PMCID: PMC7324601 DOI: 10.1038/s41598-020-67104-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/02/2020] [Indexed: 12/25/2022] Open
Abstract
EAEC is a common cause of diarrheal illness worldwide. Pathogenesis is believed to occur in the ileum and colon, where the bacteria adhere and form a robust aggregating biofilm. Among the multiple virulence factors produced by EAEC, the Pic serine protease has been implicated in bacterial colonization by virtue of its mucinolytic activity. Hence, a potential role of Pic in mucus barrier disruption during EAEC infection has been long postulated. In this study, we used human colonoids comprising goblet cells and a thick mucin barrier as an intestinal model to investigate Pic's roles during infection with EAEC. We demonstrated the ability of purified Pic, but not a protease defective Pic mutant to degrade MUC2. Western blot and confocal microscopy analysis revealed degradation of the MUC2 layer in colonoids infected with EAEC, but not with its isogenic EAECpic mutant. Wild-type and MUC2-knockdown colonoids infected with EAEC strains exposed a differential biofilm distribution, greater penetration of the mucus layer and increased colonization of the colonic epithelium by Wild-type EAEC than its isogenic Pic mutant. Higher secretion of pro-inflammatory cytokines was seen in colonoids infected with EAEC than EAECpic. Although commensal E. coli expressing Pic degraded MUC2, it did not show improved mucus layer penetration or colonization of the colonic epithelium. Our study demonstrates a role of Pic in MUC2 barrier disruption in the human intestine and shows that colonoids are a reliable system to study the interaction of pathogens with the mucus layer.
Collapse
|
48
|
Chukwu MO, Abia ALK, Ubomba-Jaswa E, Dewar JB, Obi C. Mixed Aetiology of Diarrhoea in Infants Attending Clinics in the North-West Province of South Africa: Potential for Sub-Optimal Treatment. Pathogens 2020; 9:E198. [PMID: 32155961 PMCID: PMC7157715 DOI: 10.3390/pathogens9030198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 11/28/2019] [Accepted: 12/01/2019] [Indexed: 11/17/2022] Open
Abstract
Routine diagnostic methods for the aetiologic agents of diarrhoea in most developing countries are usually not sensitive enough, leading to under-diagnosis. Thus, this study investigated possible mixed diarrhoeal aetiology by using cultures and real-time polymerase chain reactions (PCR) in children younger than four years old in the Northwest Province, South Africa. In total, 505 stool samples were collected from symptomatic and asymptomatic children who were attending three clinics and the Brits hospital in Madibeng District, between September 2016 and December 2017. Rotavirus, norovirus, Campylobacter, Arcobacter, and diarrhoeagenic Escherichia coli (DEC) were targeted. Campylobacter spp. (24.6%), Arcobacter (15.8%) and DEC (19.6%) were detected using PCR; only Campylobacter spp. (29.7%) and DEC (26.9%) were detected through the culture. Campylobacter jejuni (36%), Campylobacter coli (28%), Campylobacter upsalensis (12%), and Arcobacter butzleri (15.8%) were the only spp. of Campylobacter and Arcobacter identified. The eaeA gene (31.4%) of enteropathogenic E. coli/enterohaemorrhagic E. coli (EPEC/EHEC) was the most prevalent DEC virulence gene (VG) identified. Rotavirus and norovirus were detected at 23.4% and 20%, respectively. Mixed viral aetiology (7.3%) and the co-infection of A. butzleri and Campylobacter (49%) were recorded. A mixed bacterial-viral aetiology was observed in 0.6% of the specimens. Sensitive diagnostic procedures like PCR should be considered to provide the best treatment to children experiencing diarrhoea.
Collapse
Affiliation(s)
- Martina O. Chukwu
- Department of Life Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Corner Christiaan De wet and Pioneer Avenue, Florida Park, Roodepoort 1724, Gauteng, South Africa;
| | - Akebe Luther King Abia
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa;
| | - Eunice Ubomba-Jaswa
- Department of Biotechnology, University of Johannesburg, 37 Nind Street, Doornfontein 2094, Gauteng, South Africa;
- Water Research Commission, Lynnwood Bridge Office Park, Bloukrans Building, 4 Daventry Street, Lynnwood Manor, Pretoria 0081, South Africa
| | - John Barr Dewar
- Department of Life Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Corner Christiaan De wet and Pioneer Avenue, Florida Park, Roodepoort 1724, Gauteng, South Africa;
| | - C.L. Obi
- School of Science and Technology, Sefako Makgatho Health Science University, Ga-Rankuwa 0208, South Africa;
| |
Collapse
|
49
|
Pogreba-Brown K, Austhof E, Armstrong A, Schaefer K, Villa Zapata L, McClelland DJ, Batz MB, Kuecken M, Riddle M, Porter CK, Bazaco MC. Chronic Gastrointestinal and Joint-Related Sequelae Associated with Common Foodborne Illnesses: A Scoping Review. Foodborne Pathog Dis 2020; 17:67-86. [DOI: 10.1089/fpd.2019.2692] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Kristen Pogreba-Brown
- Epidemiology & Biostatistics Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona
| | - Erika Austhof
- Epidemiology & Biostatistics Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona
| | - Alexandra Armstrong
- Epidemiology & Biostatistics Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona
| | - Kenzie Schaefer
- Epidemiology & Biostatistics Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona
| | - Lorenzo Villa Zapata
- Epidemiology & Biostatistics Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona
| | | | | | - Maria Kuecken
- U.S. Food and Drug Administration, College Park, Maryland
| | - Mark Riddle
- Naval Medical Research Center, Silver Spring, Maryland
| | | | | |
Collapse
|
50
|
Studies on formulation of a combination heat killed immunogen from diarrheagenic Escherichia coli and Vibrio cholerae in RITARD model. Microbes Infect 2019; 21:368-376. [DOI: 10.1016/j.micinf.2019.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 01/30/2023]
|