Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Microlife ; 5: uqae005, 2024.
Article in English | MEDLINE | ID: mdl-38623411

ABSTRACT

Invasive non-typhoidal Salmonella (iNTS) disease is a serious bloodstream infection that targets immune-compromised individuals, and causes significant mortality in sub-Saharan Africa. Salmonella enterica serovar Typhimurium ST313 causes the majority of iNTS in Malawi. We performed an intensive comparative genomic analysis of 608 S. Typhimurium ST313 isolates dating between 1996 and 2018 from Blantyre, Malawi. We discovered that following the arrival of the well-characterized S. Typhimurium ST313 lineage 2 in 1999, two multidrug-resistant variants emerged in Malawi in 2006 and 2008, designated sublineages 2.2 and 2.3, respectively. The majority of S. Typhimurium isolates from human bloodstream infections in Malawi now belong to sublineages 2.2 or 2.3. To understand the emergence of the prevalent ST313 sublineage 2.2, we studied two representative strains, D23580 (lineage 2) and D37712 (sublineage 2.2). The chromosome of ST313 lineage 2 and sublineage 2.2 only differed by 29 SNPs/small indels and a 3 kb deletion of a Gifsy-2 prophage region including the sseI pseudogene. Lineage 2 and sublineage 2.2 had distinctive plasmid profiles. The transcriptome was investigated in 15 infection-relevant in vitro conditions and within macrophages. During growth in physiological conditions that do not usually trigger S. Typhimurium SPI2 gene expression, the SPI2 genes of D37712 were transcriptionally active. We identified down-regulation of flagellar genes in D37712 compared with D23580. Following phenotypic confirmation of transcriptomic differences, we discovered that sublineage 2.2 had increased fitness compared with lineage 2 during mixed growth in minimal media. We speculate that this competitive advantage is contributing to the emergence of sublineage 2.2 in Malawi.

2.
Microbiol Resour Announc ; 12(10): e0058023, 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37768056

ABSTRACT

Aeromonas caviae is an increasingly recognized etiological agent of acute gastroenteritis. Here, we report five draft genomes of A. caviae isolated from suspected cholera cases during the 2022-2023 cholera outbreak in Malawi.

3.
Lancet Psychiatry ; 8(8): 717-731, 2021 08.
Article in English | MEDLINE | ID: mdl-34115983

ABSTRACT

This scoping review of population-based epidemiological studies was done to provide background information on the prevalences and distribution of psychiatric disorders in Africa for calls to broaden diversity in psychiatric genetic studies. We searched PubMed, EMBASE, and Web of Science to retrieve relevant literature in English, French, and Portuguese from Jan 1, 1984, to Aug 18, 2020. In 36 studies from 12 African countries, the lifetime prevalence ranged from 3·3% to 9·8% for mood disorders, from 5·7% to 15·8% for anxiety disorders, from 3·7% to 13·3% for substance use disorders, and from 1·0% to 4·4% for psychotic disorders. Although the prevalence of mood and anxiety disorders appears to be lower than that observed in research outside the continent, we identified similar distributions by gender, although not by age or urbanicity. This review reveals gaps in epidemiological research on psychiatric disorders and opportunities to leverage existing epidemiological and genetic research within Africa to advance our understanding of psychiatric disorders. Studies that are methodologically comparable but diverse in geographical context are needed to advance psychiatric epidemiology and provide a foundation for understanding environmental risk in genetic studies of diverse populations globally.


Subject(s)
Anxiety Disorders/epidemiology , Mood Disorders/epidemiology , Psychotic Disorders/epidemiology , Substance-Related Disorders/epidemiology , Africa/epidemiology , Humans , Prevalence
4.
Clin Infect Dis ; 71(Suppl 2): S96-S101, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32725231

ABSTRACT

BACKGROUND: Typhoid fever remains a major source of morbidity and mortality in low-income settings. Its most feared complication is intestinal perforation. However, due to the paucity of diagnostic facilities in typhoid-endemic settings, including microbiology, histopathology, and radiology, the etiology of intestinal perforation is frequently assumed but rarely confirmed. This poses a challenge for accurately estimating burden of disease. METHODS: We recruited a prospective cohort of patients with confirmed intestinal perforation in 2016 and performed enhanced microbiological investigations (blood and tissue culture, plus tissue polymerase chain reaction [PCR] for Salmonella Typhi). In addition, we used a Poisson generalized linear model to estimate excess perforations attributed to the typhoid epidemic, using temporal trends in S. Typhi bloodstream infection and perforated abdominal viscus at Queen Elizabeth Central Hospital from 2008-2017. RESULTS: We recruited 23 patients with intraoperative findings consistent with intestinal perforation. 50% (11/22) of patients recruited were culture or PCR positive for S. Typhi. Case fatality rate from typhoid-associated intestinal perforation was substantial at 18% (2/11). Our statistical model estimates that culture-confirmed cases of typhoid fever lead to an excess of 0.046 perforations per clinical typhoid fever case (95% CI, .03-.06). We therefore estimate that typhoid fever accounts for 43% of all bowel perforation during the period of enhanced surveillance. CONCLUSIONS: The morbidity and mortality associated with typhoid abdominal perforations are high. By placing clinical outcome data from a cohort in the context of longitudinal surgical registers and bacteremia data, we describe a valuable approach to adjusting estimates of the burden of typhoid fever.


Subject(s)
Epidemics , Intestinal Perforation , Typhoid Fever , Humans , Intestinal Perforation/epidemiology , Malawi , Prospective Studies , Salmonella typhi , Typhoid Fever/complications , Typhoid Fever/epidemiology
5.
PLoS Negl Trop Dis ; 13(7): e0007539, 2019 07.
Article in English | MEDLINE | ID: mdl-31314752

ABSTRACT

Typhoid fever is endemic across sub-Saharan Africa. However, estimates of the burden of typhoid are undermined by insufficient blood volumes and lack of sensitivity of blood culture. Here, we aimed to address this limitation by exploiting pre-enrichment culture followed by PCR, alongside routine blood culture to improve typhoid case detection. We carried out a prospective diagnostic cohort study and enrolled children (aged 0-4 years) with non-specific febrile disease admitted to a tertiary hospital in Blantyre, Malawi from August 2014 to July 2016. Blood was collected for culture (BC) and real-time PCR after a pre-enrichment culture in tryptone soy broth and ox-bile. DNA was subjected to PCR for invA (Pan-Salmonella), staG (S. Typhi), and fliC (S. Typhimurium) genes. A positive PCR was defined as invA plus either staG or fliC (CT<29). IgM and IgG ELISA against four S. Typhi antigens was also performed. In total, 643 children (median age 1.3 years) with nonspecific febrile disease were enrolled; 31 (4.8%) were BC positive for Salmonella (n = 13 S. Typhi, n = 16 S. Typhimurium, and n = 2 S. Enteritidis). Pre-enrichment culture of blood followed by PCR identified a further 8 S. Typhi and 15 S. Typhimurium positive children. IgM and IgG titres to the S. Typhi antigen STY1498 (haemolysin) were significantly higher in children that were PCR positive but blood culture negative compared to febrile children with all other non-typhoid illnesses. The addition of pre-enrichment culture and PCR increased the case ascertainment of invasive Salmonella disease in children by 62-94%. These data support recent burden estimates that highlight the insensitivity of blood cultures and support the targeting of pre-school children for typhoid vaccine prevention in Africa. Blood culture with real-time PCR following pre-enrichment should be used to further refine estimates of vaccine effectiveness in typhoid vaccine trials.


Subject(s)
Bacterial Load , Cost of Illness , Fever/microbiology , Typhoid Fever/epidemiology , Antigens, Bacterial/genetics , Blood Culture , Child, Preschool , Female , Fever/epidemiology , Hospitalization , Humans , Infant , Infant, Newborn , Malawi/epidemiology , Male , Prospective Studies , Salmonella typhi/genetics , Typhoid Fever/blood , Typhoid Fever/diagnosis
6.
Wellcome Open Res ; 4: 74, 2019.
Article in English | MEDLINE | ID: mdl-31231691

ABSTRACT

Background: Salmonella Typhimurium ST313 exhibits signatures of adaptation to invasive human infection, including higher resistance to humoral immune responses than gastrointestinal isolates. Full resistance to antibody-mediated complement killing (serum resistance) among nontyphoidal Salmonellae is uncommon, but selection of highly resistant strains could compromise vaccine-induced antibody immunity. Here, we address the hypothesis that serum resistance is due to a distinct genotype or transcriptome response in S. Typhimurium ST313. Methods: Six S. Typhimurium ST313 bloodstream isolates, three of which were antibody resistant, were studied. Genomic content (single nucleotide polymorphisms and larger chromosomal modifications) of the strains was determined by Illumina and PACBIO sequencing, and functionally characterized using RNA-seq, transposon directed insertion site sequencing (TraDIS), targeted gene deletion and transfer of selected point mutations in an attempt to identify features associated with serum resistance.   Results: Sequence polymorphisms in genes from strains with atypical serum susceptibility when transferred from strains that were highly resistant or susceptible to a strain that exhibited intermediate susceptibility did not significantly alter serum killing phenotype. No large chromosomal modifications typified serum resistance or susceptibility. Genes required for resistance to serum identified by TraDIS and RNA-seq included those involved in exopolysaccharide synthesis, iron scavenging and metabolism. Most of the down-regulated genes were associated with membrane proteins. Resistant and susceptible strains had distinct transcriptional responses to serum, particularly related to genes responsible for polysaccharide biosynthesis. There was higher upregulation of wca locus genes, involved in the biosynthesis of colanic acid exopolysaccharide, in susceptible strains and increased expression of fepE, a regulator of very long-chain lipopolysaccharide in resistant strains. Conclusion: Clinical isolates of S. Typhimurium ST313 exhibit distinct antibody susceptibility phenotypes that may be associated with changes in gene expression on exposure to serum.

7.
J Antimicrob Chemother ; 74(5): 1223-1232, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30778540

ABSTRACT

OBJECTIVES: ESBL-producing Klebsiella pneumoniae (KPN) pose a major threat to human health globally. We carried out a WGS study to understand the genetic background of ESBL-producing KPN in Malawi and place them in the context of other global isolates. METHODS: We sequenced genomes of 72 invasive and carriage KPN isolates collected from patients admitted to Queen Elizabeth Central Hospital, Blantyre, Malawi. We performed phylogenetic and population structure analyses on these and previously published genomes from Kenya (n = 66) and from outside sub-Saharan Africa (n = 67). We screened for presence of antimicrobial resistance (AMR) genetic determinants and carried out association analyses by genomic sequence cluster, AMR phenotype and time. RESULTS: Malawian isolates fit within the global population structure of KPN, clustering into the major lineages of KpI, KpII and KpIII. KpI isolates from Malawi were more related to those from Kenya, with both collections exhibiting more clonality than isolates from the rest of the world. We identified multiple ESBL genes, including blaCTX-M-15, several blaSHV, blaTEM-63 and blaOXA-10, and other AMR genes, across diverse lineages of the KPN isolates from Malawi. No carbapenem resistance genes were detected; however, we detected IncFII and IncFIB plasmids that were similar to the carbapenem resistance-associated plasmid pNDM-mar. CONCLUSIONS: There are multiple ESBL genes across diverse KPN lineages in Malawi and plasmids in circulation that are capable of carrying carbapenem resistance. Unless appropriate interventions are rapidly put in place, these may lead to a high burden of locally untreatable infection in vulnerable populations.


Subject(s)
Genome, Bacterial , Genomics , Klebsiella Infections/microbiology , Klebsiella pneumoniae/classification , Klebsiella pneumoniae/genetics , beta-Lactamases/genetics , Anti-Bacterial Agents/pharmacology , Computational Biology/methods , Drug Resistance, Multiple, Bacterial , Genetic Variation , Genomics/methods , Humans , Klebsiella pneumoniae/isolation & purification , Malawi , Microbial Sensitivity Tests , Phylogeny
8.
Nat Commun ; 9(1): 1014, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523850

ABSTRACT

Nontyphoidal Salmonella (NTS) is a major cause of bacteraemia in Africa. The disease typically affects HIV-infected individuals and young children, causing substantial morbidity and mortality. Here we present a genome-wide association study (180 cases, 2677 controls) and replication analysis of NTS bacteraemia in Kenyan and Malawian children. We identify a locus in STAT4, rs13390936, associated with NTS bacteraemia. rs13390936 is a context-specific expression quantitative trait locus for STAT4 RNA expression, and individuals carrying the NTS-risk genotype demonstrate decreased interferon-γ (IFNγ) production in stimulated natural killer cells, and decreased circulating IFNγ concentrations during acute NTS bacteraemia. The NTS-risk allele at rs13390936 is associated with protection against a range of autoimmune diseases. These data implicate interleukin-12-dependent IFNγ-mediated immunity as a determinant of invasive NTS disease in African children, and highlight the shared genetic architecture of infectious and autoimmune disease.


Subject(s)
Autoimmune Diseases/genetics , Bacteremia/epidemiology , Genetic Predisposition to Disease , STAT4 Transcription Factor/genetics , Salmonella Infections/epidemiology , Salmonella/pathogenicity , Adolescent , Alleles , Autoimmune Diseases/epidemiology , Autoimmune Diseases/immunology , Autoimmune Diseases/microbiology , Bacteremia/genetics , Bacteremia/immunology , Bacteremia/microbiology , Case-Control Studies , Child , Child, Preschool , Female , Follow-Up Studies , Genome-Wide Association Study , Genotype , Humans , Immunity, Cellular/genetics , Infant , Infant, Newborn , Interferon-gamma/blood , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-12/immunology , Interleukin-12/metabolism , Kenya/epidemiology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Malawi/epidemiology , Male , Polymorphism, Single Nucleotide , Quantitative Trait Loci/genetics , Risk Factors , Salmonella/isolation & purification , Salmonella Infections/genetics , Salmonella Infections/immunology , Salmonella Infections/microbiology
9.
PLoS Negl Trop Dis ; 11(12): e0006027, 2017 12.
Article in English | MEDLINE | ID: mdl-29216183

ABSTRACT

Nontyphoidal Salmonellae commonly cause invasive disease in African children that is often fatal. The clinical diagnosis of these infections is hampered by the absence of a clear clinical syndrome. Drug resistance means that empirical antibiotic therapy is often ineffective and currently no vaccine is available. The study objective was to identify risk factors for mortality among children presenting to hospital with invasive Salmonella disease in Africa. We conducted a prospective study enrolling consecutive children with microbiologically-confirmed invasive Salmonella disease admitted to Queen Elizabeth Central Hospital, Blantyre, in 2006. Data on clinical presentation, co-morbidities and outcome were used to identify children at risk of inpatient mortality through logistic-regression modeling. Over one calendar year, 263 consecutive children presented with invasive Salmonella disease. Median age was 16 months (range 0-15 years) and 52/256 children (20%; 95%CI 15-25%) died. Nontyphoidal serovars caused 248/263 (94%) of cases. 211/259 (81%) of isolates were multi-drug resistant. 251/263 children presented with bacteremia, 6 with meningitis and 6 with both. Respiratory symptoms were present in 184/240 (77%; 95%CI 71-82%), 123/240 (51%; 95%CI 45-58%) had gastrointestinal symptoms and 101/240 (42%; 95%CI 36-49%) had an overlapping clinical syndrome. Presentation at <7 months (OR 10.0; 95%CI 2.8-35.1), dyspnea (OR 4.2; 95%CI 1.5-12.0) and HIV infection (OR 3.3; 95%CI 1.1-10.2) were independent risk factors for inpatient mortality. Invasive Salmonella disease in Malawi is characterized by high mortality and prevalence of multi-drug resistant isolates, along with non-specific presentation. Young infants, children with dyspnea and HIV-infected children bear a disproportionate burden of the Salmonella-associated mortality in Malawi. Strategies to improve prevention, diagnosis and management of invasive Salmonella disease should be targeted at these children.


Subject(s)
Bacteremia/epidemiology , HIV Infections/complications , Meningitis, Bacterial/epidemiology , Salmonella Infections/epidemiology , Salmonella/immunology , Adolescent , Bacteremia/etiology , Bacteremia/microbiology , Bacteremia/mortality , Child , Child, Preschool , Drug Resistance, Multiple, Bacterial , Female , Humans , Infant , Logistic Models , Malawi/epidemiology , Male , Meningitis, Bacterial/etiology , Meningitis, Bacterial/microbiology , Meningitis, Bacterial/mortality , Prevalence , Prospective Studies , Risk Factors , Salmonella/isolation & purification , Salmonella Infections/etiology , Salmonella Infections/microbiology , Salmonella Infections/mortality , Serogroup
10.
Lancet Infect Dis ; 17(10): 1042-1052, 2017 10.
Article in English | MEDLINE | ID: mdl-28818544

ABSTRACT

BACKGROUND: Bacterial bloodstream infection is a common cause of morbidity and mortality in sub-Saharan Africa, yet few facilities are able to maintain long-term surveillance. The Malawi-Liverpool-Wellcome Trust Clinical Research Programme has done sentinel surveillance of bacteraemia since 1998. We report long-term trends in bloodstream infection and antimicrobial resistance from this surveillance. METHODS: In this surveillance study, we analysed blood cultures that were routinely taken from adult and paediatric patients with fever or suspicion of sepsis admitted to Queen Elizabeth Central Hospital, Blantyre, Malawi from 1998 to 2016. The hospital served an urban population of 920 000 in 2016, with 1000 beds, although occupancy often exceeds capacity. The hospital admits about 10 000 adults and 30 000 children each year. Antimicrobial susceptibility tests were done by the disc diffusion method according to British Society of Antimicrobial Chemotherapy guidelines. We used the Cochran-Armitage test for trend to examine trends in rates of antimicrobial resistance, and negative binomial regression to examine trends in icidence of bloodstream infection over time. FINDINGS: Between Jan 1, 1998, and Dec 31, 2016, we isolated 29 183 pathogens from 194 539 blood cultures. Pathogen detection decreased significantly from 327·1/100 000 in 1998 to 120·2/100 000 in 2016 (p<0·0001). 13 366 (51·1%) of 26 174 bacterial isolates were resistant to the Malawian first-line antibiotics amoxicillin or penicillin, chloramphenicol, and co-trimoxazole; 68·3% of Gram-negative and 6·6% of Gram-positive pathogens. The proportions of non-Salmonella Enterobacteriaceae with extended spectrum beta-lactamase (ESBL) or fluoroquinolone resistance rose significantly after 2003 to 61·9% in 2016 (p<0·0001). Between 2003 and 2016, ESBL resistance rose from 0·7% to 30·3% in Escherichia coli, from 11·8% to 90·5% in Klebsiella spp and from 30·4% to 71·9% in other Enterobacteriaceae. Similarly, resistance to ciprofloxacin rose from 2·5% to 31·1% in E coli, from 1·7% to 70·2% in Klebsiella spp and from 5·9% to 68·8% in other Enterobacteriaceae. By contrast, more than 92·0% of common Gram-positive pathogens remain susceptible to either penicillin or chloramphenicol. Meticillin-resistant Staphylococcus aureus (MRSA) was first reported in 1998 at 7·7% and represented 18·4% of S aureus isolates in 2016. INTERPRETATION: The rapid expansion of ESBL and fluoroquinolone resistance among common Gram-negative pathogens, and the emergence of MRSA, highlight the growing challenge of bloodstream infections that are effectively impossible to treat in this resource-limited setting. FUNDING: Wellcome Trust, H3ABionet, Southern Africa Consortium for Research Excellence (SACORE).


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Hospitals , Sepsis/microbiology , Adult , Child , Drug Resistance, Bacterial , Humans , Incidence , Malawi/epidemiology , Population Surveillance , Retrospective Studies , Sepsis/epidemiology , Time Factors
11.
Vaccine ; 35(35 Pt B): 4594-4602, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28711389

ABSTRACT

INTRODUCTION: Pneumococcal infections cause a high death toll in Sub Saharan Africa (SSA) but the recently rolled out pneumococcal conjugate vaccines (PCV) will reduce the disease burden. To better understand the population impact of these vaccines, comprehensive analysis of large collections of pneumococcal isolates sampled prior to vaccination is required. Here we present a population genomic study of the invasive pneumococcal isolates sampled before the implementation of PCV13 in Malawi. MATERIALS AND METHODS: We retrospectively sampled and whole genome sequenced 585 invasive isolates from 2004 to 2010. We determine the pneumococcal population genetic structure and assessed serotype prevalence, antibiotic resistance rates, and the occurrence of serotype switching. RESULTS: Population structure analysis revealed 22 genetically distinct sequence clusters (SCs), which consisted of closely related isolates. Serotype 1 (ST217), a vaccine-associated serotype in clade SC2, showed highest prevalence (19.3%), and was associated with the highest MDR rate (81.9%) followed by serotype 12F, a non-vaccine serotype in clade SC10 with an MDR rate of 57.9%. Prevalence of serotypes was stable prior to vaccination although there was an increase in the PMEN19 clone, serotype 5 ST289, in clade SC1 in 2010 suggesting a potential undetected local outbreak. Coalescent analysis revealed recent emergence of the SCs and there was evidence of natural capsule switching in the absence of vaccine induced selection pressure. Furthermore, majority of the highly prevalent capsule-switched isolates were associated with acquisition of vaccine-targeted capsules. CONCLUSIONS: This study provides descriptions of capsule-switched serotypes and serotypes with potential to cause serotype replacement post-vaccination such as 12F. Continued surveillance is critical to monitor these serotypes and antibiotic resistance in order to design better infection prevention and control measures such as inclusion of emerging replacement serotypes in future conjugate vaccines.


Subject(s)
Bacterial Capsules/genetics , Pneumococcal Infections/microbiology , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/genetics , Adolescent , Adult , Bacterial Capsules/physiology , Child , Child, Preschool , Drug Resistance, Multiple, Bacterial , Evolution, Molecular , Female , Genome, Bacterial , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Malawi/epidemiology , Male , Pneumococcal Infections/epidemiology , Prevalence , Retrospective Studies , Serogroup , Serotyping , Streptococcus pneumoniae/isolation & purification , Streptococcus pneumoniae/physiology , Vaccination , Vaccines, Conjugate/immunology , Young Adult
12.
Clin Vaccine Immunol ; 24(7)2017 Jul.
Article in English | MEDLINE | ID: mdl-28515136

ABSTRACT

Invasive nontyphoidal Salmonella (iNTS) infections are commonly associated with Plasmodium falciparum infections, but the immunologic basis for this linkage is poorly understood. We hypothesized that P. falciparum infection compromises the humoral and cellular immunity of the host to NTS, which increases the susceptibility of the host to iNTS infection. We prospectively recruited children aged between 6 and 60 months at a Community Health Centre in Blantyre, Malawi, and allocated them to the following groups; febrile with uncomplicated malaria, febrile malaria negative, and nonfebrile malaria negative. Levels of Salmonella enterica serovar Typhimurium-specific serum bactericidal activity (SBA) and whole-blood bactericidal activity (WBBA), complement C3 deposition, and neutrophil respiratory burst activity (NRBA) were measured. Levels of SBA with respect to S Typhimurium were reduced in febrile P. falciparum-infected children (median, -0.20 log10 [interquartile range {IQR}, -1.85, 0.32]) compared to nonfebrile malaria-negative children (median, -1.42 log10 [IQR, -2.0, -0.47], P = 0.052). In relation to SBA, C3 deposition on S Typhimurium was significantly reduced in febrile P. falciparum-infected children (median, 7.5% [IQR, 4.1, 15.0]) compared to nonfebrile malaria-negative children (median, 29% [IQR, 11.8, 48.0], P = 0.048). WBBA with respect to S Typhimurium was significantly reduced in febrile P. falciparum-infected children (median, 0.25 log10 [IQR, -0.73, 1.13], P = 0.0001) compared to nonfebrile malaria-negative children (median, -1.0 log10 [IQR, -1.68, -0.16]). In relation to WBBA, S Typhimurium-specific NRBA was reduced in febrile P. falciparum-infected children (median, 8.8% [IQR, 3.7, 20], P = 0.0001) compared to nonfebrile malaria-negative children (median, 40.5% [IQR, 33, 65.8]). P. falciparum infection impairs humoral and cellular immunity to S Typhimurium in children during malaria episodes, which may explain the increased risk of iNTS observed in children from settings of malaria endemicity. The mechanisms underlying humoral immunity impairment are incompletely understood and should be explored further.


Subject(s)
Blood Bactericidal Activity , Disease Susceptibility , Immunity, Cellular , Immunity, Humoral , Malaria, Falciparum/complications , Salmonella Infections/immunology , Salmonella Vaccines/immunology , Child, Preschool , Complement System Proteins/metabolism , Female , Humans , Infant , Malawi , Male , Neutrophils/immunology , Prospective Studies , Respiratory Burst , Salmonella Infections/epidemiology , Salmonella typhimurium/immunology
13.
J Antimicrob Chemother ; 72(6): 1602-1609, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28333330

ABSTRACT

Objectives: Efforts to treat Escherichia coli infections are increasingly being compromised by the rapid, global spread of antimicrobial resistance (AMR). Whilst AMR in E. coli has been extensively investigated in resource-rich settings, in sub-Saharan Africa molecular patterns of AMR are not well described. In this study, we have begun to explore the population structure and molecular determinants of AMR amongst E. coli isolates from Malawi. Methods: Ninety-four E. coli isolates from patients admitted to Queen's Hospital, Malawi, were whole-genome sequenced. The isolates were selected on the basis of diversity of phenotypic resistance profiles and clinical source of isolation (blood, CSF and rectal swab). Sequence data were analysed using comparative genomics and phylogenetics. Results: Our results revealed the presence of five clades, which were strongly associated with E. coli phylogroups A, B1, B2, D and F. We identified 43 multilocus STs, of which ST131 (14.9%) and ST12 (9.6%) were the most common. We identified 25 AMR genes. The most common ESBL gene was bla CTX-M-15 and it was present in all five phylogroups and 11 STs, and most commonly detected in ST391 (4/4 isolates), ST648 (3/3 isolates) and ST131 [3/14 (21.4%) isolates]. Conclusions: This study has revealed a high diversity of lineages associated with AMR, including ESBL and fluoroquinolone resistance, in Malawi. The data highlight the value of longitudinal bacteraemia surveillance coupled with detailed molecular epidemiology in all settings, including low-income settings, in describing the global epidemiology of ESBL resistance.


Subject(s)
Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli Infections/microbiology , Escherichia coli/drug effects , Escherichia coli/genetics , beta-Lactam Resistance/genetics , beta-Lactamases/genetics , Adolescent , Adult , Child , Child, Preschool , Chloramphenicol/pharmacology , Escherichia coli/isolation & purification , Escherichia coli Infections/epidemiology , Female , Genes, Bacterial , Genetic Variation , Genomics , Humans , Malawi/epidemiology , Male , Microbial Sensitivity Tests , Multilocus Sequence Typing , Phylogeny , Urban Population/statistics & numerical data , Young Adult
14.
Clin Vaccine Immunol ; 24(4)2017 Apr.
Article in English | MEDLINE | ID: mdl-28122790

ABSTRACT

Proinflammatory cytokines are involved in clearance of Plasmodium falciparum, and very high levels of these cytokines have been implicated in the pathogenesis of severe malaria. In order to determine how cytokines vary with disease severity and syndrome, we enrolled Malawian children presenting with cerebral malaria (CM), severe malarial anemia (SMA), and uncomplicated malaria (UCM) and healthy controls. We analyzed serum cytokine concentrations in acute infection and in convalescence. With the exception of interleukin 5 (IL-5), cytokine concentrations were highest in acute CM, followed by SMA, and were only mildly elevated in UCM. Cytokine concentrations had fallen to control levels when remeasured at 1 month of convalescence in all three clinical malaria groups. Ratios of IL-10 to tumor necrosis factor alpha (TNF-α) and of IL-10 to IL-6 followed a similar pattern. Children presenting with acute CM had significantly higher concentrations of TNF-α (P < 0.001), interferon gamma (IFN-γ) (P = 0.0019), IL-2 (P = 0.0004), IL-6 (P < 0.001), IL-8 (P < 0.001), and IL-10 (P < 0.001) in sera than healthy controls. Patients with acute CM had significantly higher concentrations of IL-6 (P < 0.001) and IL-10 (P = 0.0003) than those presenting with acute SMA. Our findings are consistent with the concept that high levels of proinflammatory cytokines, despite high levels of the anti-inflammatory cytokine IL-10, could contribute to the pathogenesis of CM.


Subject(s)
Cytokines/blood , Malaria, Falciparum/immunology , Child , Child, Preschool , Female , Humans , Infant , Male , Serum/chemistry
15.
Clin Vaccine Immunol ; 23(7): 601-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27170644

ABSTRACT

Nontyphoidal Salmonella is a leading cause of sepsis in African children. Cytokine responses are central to the pathophysiology of sepsis and predict sepsis outcome in other settings. In this study, we investigated cytokine responses to invasive nontyphoidal Salmonella (iNTS) disease in Malawian children. We determined serum concentrations of 48 cytokines with multiplexed immunoassays in Malawian children during acute iNTS disease (n = 111) and in convalescence (n = 77). Principal component analysis and logistic regression were used to identify cytokine signatures of acute iNTS disease. We further investigated whether these responses are altered by HIV coinfection or severe malnutrition and whether cytokine responses predict inpatient mortality. Cytokine changes in acute iNTS disease were associated with two distinct cytokine signatures. The first is characterized by increased concentrations of mediators known to be associated with macrophage function, and the second is characterized by raised pro- and anti-inflammatory cytokines typical of responses reported in sepsis secondary to diverse pathogens. These cytokine responses were largely unaltered by either severe malnutrition or HIV coinfection. Children with fatal disease had a distinctive cytokine profile, characterized by raised mediators known to be associated with neutrophil function. In conclusion, cytokine responses to acute iNTS infection in Malawian children are reflective of both the cytokine storm typical of sepsis secondary to diverse pathogens and the intramacrophage replicative niche of NTS. The cytokine profile predictive of fatal disease supports a key role of neutrophils in the pathogenesis of NTS sepsis.


Subject(s)
Cytokines/blood , Salmonella Infections/mortality , Salmonella Infections/pathology , Sepsis/mortality , Sepsis/pathology , Adolescent , Child , Child, Preschool , Female , HIV Infections/complications , Humans , Infant , Infant, Newborn , Macrophages/immunology , Malawi , Male , Malnutrition/complications , Neutrophils/immunology , Survival Analysis
16.
PLoS Negl Trop Dis ; 10(4): e0004604, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27057743

ABSTRACT

BACKGROUND: Nontyphoidal strains of Salmonella are a leading cause of death among HIV-infected Africans. Antibody-induced complement-mediated killing protects healthy Africans against Salmonella, but increased levels of anti-lipopolysaccharide (LPS) antibodies in some HIV-infected African adults block this killing. The objective was to understand how these high levels of anti-LPS antibodies interfere with the killing of Salmonella. METHODOLOGY/PRINCIPAL FINDINGS: Sera and affinity-purified antibodies from African HIV-infected adults that failed to kill invasive S. Typhimurium D23580 were compared to sera from HIV-uninfected and HIV-infected subjects with bactericidal activity. The failure of sera from certain HIV-infected subjects to kill Salmonella was found to be due to an inherent inhibitory effect of anti-LPS antibodies. This inhibition was concentration-dependent and strongly associated with IgA and IgG2 anti-LPS antibodies (p<0.0001 for both). IgG anti-LPS antibodies, from sera of HIV-infected individuals that inhibit killing at high concentration, induced killing when diluted. Conversely, IgG, from sera of HIV-uninfected adults that induce killing, inhibited killing when concentrated. IgM anti-LPS antibodies from all subjects also induced Salmonella killing. Finally, the inhibitory effect of high concentrations of anti-LPS antibodies is seen with IgM as well as IgG and IgA. No correlation was found between affinity or avidity, or complement deposition or consumption, and inhibition of killing. CONCLUSION/SIGNIFICANCE: IgG and IgM classes of anti-S. Typhimurium LPS antibodies from HIV-infected and HIV-uninfected individuals are bactericidal, while at very high concentrations, anti-LPS antibodies of all classes inhibit in vitro killing of Salmonella. This could be due to a variety of mechanisms relating to the poor ability of IgA and IgG2 to activate complement, and deposition of complement at sites where it cannot insert in the bacterial membrane. Vaccine trials are required to understand the significance of lack of in vitro killing by anti-LPS antibodies from a minority of HIV-infected individuals with impaired immune homeostasis.


Subject(s)
Antibodies, Bacterial/blood , HIV Infections/immunology , Salmonella Infections/immunology , Salmonella/immunology , Africa , Immunocompromised Host , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Lipopolysaccharides/immunology , Serum Bactericidal Antibody Assay , Serum Bactericidal Test
17.
mBio ; 7(2): e02265, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26933058

ABSTRACT

UNLABELLED: Nontyphoidal Salmonella enterica serovar Typhimurium is a frequent cause of bloodstream infections in children and HIV-infected adults in sub-Saharan Africa. Most isolates from African patients with bacteremia belong to a single sequence type, ST313, which is genetically distinct from gastroenteritis-associated ST19 strains, such as 14028s and SL1344. Some studies suggest that the rapid spread of ST313 across sub-Saharan Africa has been facilitated by anthroponotic (person-to-person) transmission, eliminating the need for Salmonella survival outside the host. While these studies have not ruled out zoonotic or other means of transmission, the anthroponotic hypothesis is supported by evidence of extensive genomic decay, a hallmark of host adaptation, in the sequenced ST313 strain D23580. We have identified and demonstrated 2 loss-of-function mutations in D23580, not present in the ST19 strain 14028s, that impair multicellular stress resistance associated with survival outside the host. These mutations result in inactivation of the KatE stationary-phase catalase that protects high-density bacterial communities from oxidative stress and the BcsG cellulose biosynthetic enzyme required for the RDAR (red, dry, and rough) colonial phenotype. However, we found that like 14028s, D23580 is able to elicit an acute inflammatory response and cause enteritis in mice and rhesus macaque monkeys. Collectively, these observations suggest that African S. Typhimurium ST313 strain D23580 is becoming adapted to an anthroponotic mode of transmission while retaining the ability to infect and cause enteritis in multiple host species. IMPORTANCE: The last 3 decades have witnessed an epidemic of invasive nontyphoidal Salmonella infections in sub-Saharan Africa. Genomic analysis and clinical observations suggest that the Salmonella strains responsible for these infections are evolving to become more typhoid-like with regard to patterns of transmission and virulence. This study shows that a prototypical African nontyphoidal Salmonella strain has lost traits required for environmental stress resistance, consistent with an adaptation to a human-to-human mode of transmission. However, in contrast to predictions, the strain remains capable of causing acute inflammation in the mammalian intestine. This suggests that the systemic clinical presentation of invasive nontyphoidal Salmonella infections in Africa reflects the immune status of infected hosts rather than intrinsic differences in the virulence of African Salmonella strains. Our study provides important new insights into the evolution of host adaptation in bacterial pathogens.


Subject(s)
Adaptation, Biological , Salmonella Infections/microbiology , Salmonella typhimurium/enzymology , Salmonella typhimurium/physiology , Stress, Physiological , Africa South of the Sahara/epidemiology , Animals , Catalase/genetics , Catalase/metabolism , Disease Models, Animal , Epidemics , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Humans , Macaca mulatta , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Salmonella Infections/epidemiology , Salmonella typhimurium/genetics , Salmonella typhimurium/isolation & purification
18.
Clin Vaccine Immunol ; 23(2): 95-103, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26581890

ABSTRACT

Lymphocytes are implicated in immunity and pathogenesis of severe malaria. Since lymphocyte subsets vary with age, assessment of their contribution to different etiologies can be difficult. We immunophenotyped peripheral blood from Malawian children presenting with cerebral malaria, severe malarial anemia, and uncomplicated malaria (n = 113) and healthy aparasitemic children (n = 42) in Blantyre, Malawi, and investigated lymphocyte subset counts, activation, and memory status. Children with cerebral malaria were older than those with severe malarial anemia. We found panlymphopenia in children presenting with cerebral malaria (median lymphocyte count, 2,100/µl) and uncomplicated malaria (3,700/µl), which was corrected in convalescence and was absent in severe malarial anemia (5,950/µl). Median percentages of activated CD69(+) NK (73%) and γδ T (60%) cells were higher in cerebral malaria than in other malaria types. Median ratios of memory to naive CD4(+) lymphocytes were higher in cerebral malaria than in uncomplicated malaria and low in severe malarial anemia. The polarized lymphocyte subset profiles of different forms of severe malaria are independent of age. In conclusion, among Malawian children cerebral malaria is characterized by lymphocyte activation and increased memory cells, consistent with immune priming. In contrast, there are reduced memory cells and less activation in severe malaria anemia. Further studies are required to understand whether these immunological profiles indicate predisposition of some children to one or another form of severe malaria.


Subject(s)
Immunologic Memory , Lymphocyte Activation , Lymphocyte Subsets/immunology , Malaria, Cerebral/immunology , Malaria, Falciparum/immunology , T-Lymphocyte Subsets/immunology , Anemia/epidemiology , Anemia/immunology , Anemia/mortality , Anemia/parasitology , Child , Child, Preschool , Disease Susceptibility , Female , Humans , Infant , Lymphocyte Count , Malaria, Cerebral/epidemiology , Malaria, Cerebral/mortality , Malaria, Cerebral/parasitology , Malaria, Falciparum/complications , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Malawi/epidemiology , Male
20.
Nat Genet ; 44(11): 1215-21, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23023330

ABSTRACT

A highly invasive form of non-typhoidal Salmonella (iNTS) disease has recently been documented in many countries in sub-Saharan Africa. The most common Salmonella enterica serovar causing this disease is Typhimurium (Salmonella Typhimurium). We applied whole-genome sequence-based phylogenetic methods to define the population structure of sub-Saharan African invasive Salmonella Typhimurium isolates and compared these to global Salmonella Typhimurium populations. Notably, the vast majority of sub-Saharan invasive Salmonella Typhimurium isolates fell within two closely related, highly clustered phylogenetic lineages that we estimate emerged independently ∼52 and ∼35 years ago in close temporal association with the current HIV pandemic. Clonal replacement of isolates from lineage I by those from lineage II was potentially influenced by the use of chloramphenicol for the treatment of iNTS disease. Our analysis suggests that iNTS disease is in part an epidemic in sub-Saharan Africa caused by highly related Salmonella Typhimurium lineages that may have occupied new niches associated with a compromised human population and antibiotic treatment.


Subject(s)
Drug Resistance, Multiple, Bacterial/genetics , Phylogeny , Salmonella Infections , Salmonella typhimurium , Africa South of the Sahara/epidemiology , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , Evolution, Molecular , HIV Infections/epidemiology , Humans , Phylogeography , Polymorphism, Single Nucleotide , Salmonella Infections/epidemiology , Salmonella Infections/genetics , Salmonella typhimurium/classification , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...