Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
Add more filters

Publication year range
1.
Brief Bioinform ; 24(2)2023 03 19.
Article in English | MEDLINE | ID: mdl-36804804

ABSTRACT

Recent technological and computational advances have made metagenomic assembly a viable approach to achieving high-resolution views of complex microbial communities. In previous benchmarking, short-read (SR) metagenomic assemblers had the highest accuracy, long-read (LR) assemblers generated the most contiguous sequences and hybrid (HY) assemblers balanced length and accuracy. However, no assessments have specifically compared the performance of these assemblers on low-abundance species, which include clinically relevant organisms in the gut. We generated semi-synthetic LR and SR datasets by spiking small and increasing amounts of Escherichia coli isolate reads into fecal metagenomes and, using different assemblers, examined E. coli contigs and the presence of antibiotic resistance genes (ARGs). For ARG assembly, although SR assemblers recovered more ARGs with high accuracy, even at low coverages, LR assemblies allowed for the placement of ARGs within longer, E. coli-specific contigs, thus pinpointing their taxonomic origin. HY assemblies identified resistance genes with high accuracy and had lower contiguity than LR assemblies. Each assembler type's strengths were maintained even when our isolate was spiked in with a competing strain, which fragmented and reduced the accuracy of all assemblies. For strain characterization and determining gene context, LR assembly is optimal, while for base-accurate gene identification, SR assemblers outperform other options. HY assembly offers contiguity and base accuracy, but requires generating data on multiple platforms, and may suffer high misassembly rates when strain diversity exists. Our results highlight the trade-offs associated with each approach for recovering low-abundance taxa, and that the optimal approach is goal-dependent.


Subject(s)
Metagenome , Microbiota , Sequence Analysis, DNA/methods , Escherichia coli/genetics , Microbiota/genetics , Metagenomics/methods , High-Throughput Nucleotide Sequencing/methods
2.
Clin Infect Dis ; 78(1): 31-39, 2024 01 25.
Article in English | MEDLINE | ID: mdl-37633257

ABSTRACT

BACKGROUND: The clinical and microbial factors associated with Klebsiella pneumoniae bloodstream infections (BSIs) are not well characterized. Prior studies have focused on highly resistant or hypervirulent isolates, limiting our understanding of K. pneumoniae strains that commonly cause BSI. We performed a record review and whole-genome sequencing to investigate the clinical characteristics, bacterial diversity, determinants of antimicrobial resistance, and risk factors for in-hospital death in a cohort of patients with K. pneumoniae BSI. METHODS: We identified 562 patients at Massachusetts General Hospital with K. pneumoniae BSIs between 2016 and 2022. We collected data on comorbid conditions, infection source, clinical outcomes, and antibiotic resistance and performed whole-genome sequencing on 108 sequential BSI isolates from 2021 to 2022. RESULTS: Intra-abdominal infection was the most common source of infection accounting for 34% of all BSIs. A respiratory tract source accounted for 6% of BSIs but was associated with a higher in-hospital mortality rate (adjusted odds ratio, 5.4 [95% confidence interval, 2.2-12.8]; P < .001 for comparison with other sources). Resistance to the first antibiotic prescribed was also associated with a higher risk of death (adjusted odds ratio, 5.2 [95% confidence interval, 2.2-12.4]; P < .001). BSI isolates were genetically diverse, and no clusters of epidemiologically and genetically linked cases were observed. Virulence factors associated with invasiveness were observed at a low prevalence, although an unexpected association between O-antigen type and the source of infection was found. CONCLUSIONS: These observations demonstrate the versatility of K. pneumoniae as an opportunistic pathogen and highlight the need for new approaches for surveillance and the rapid identification of patients with invasive antimicrobial-resistant K. pneumoniae infection.


Subject(s)
Bacteremia , Cross Infection , Klebsiella Infections , Sepsis , Humans , Klebsiella pneumoniae , Cross Infection/epidemiology , Hospital Mortality , Bacteremia/microbiology , Klebsiella Infections/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Sepsis/drug therapy , Genomics
3.
Ann Intern Med ; 176(9): ITC129-ITC144, 2023 09.
Article in English | MEDLINE | ID: mdl-37696033

ABSTRACT

International travel can cause new illness or exacerbate existing conditions. Because primary care providers are frequent sources of health advice to travelers, they should be familiar with destination-specific disease risks, be knowledgeable about travel and routine vaccines, be prepared to prescribe chemoprophylaxis and self-treatment regimens, and be aware of travel medicine resources. Primary care providers should recognize travelers who would benefit from referral to a specialized travel clinic for evaluation. Those requiring yellow fever vaccination, immunocompromised hosts, pregnant persons, persons with multiple comorbid conditions, or travelers with complex itineraries may warrant specialty referral.


Subject(s)
Medicine , Travel Medicine , Female , Pregnancy , Humans , Ambulatory Care Facilities , Awareness , Chemoprevention
4.
Clin Infect Dis ; 76(5): 950-956, 2023 03 04.
Article in English | MEDLINE | ID: mdl-36048507

ABSTRACT

The earth is rapidly warming, driven by increasing atmospheric carbon dioxide and other gases that result primarily from fossil fuel combustion. In addition to causing arctic ice melting and extreme weather events, climatologic factors are linked strongly to the transmission of many infectious diseases. Changes in the prevalence of infectious diseases not only reflect the impacts of temperature, humidity, and other weather-related phenomena on pathogens, vectors, and animal hosts but are also part of a complex of social and environmental factors that will be affected by climate change, including land use, migration, and vector control. Vector- and waterborne diseases and coccidioidomycosis are all likely to be affected by a warming planet; there is also potential for climate-driven impacts on emerging infectious diseases and antimicrobial resistance. Additional resources for surveillance and public health activities are urgently needed, as well as systematic education of clinicians on the health impacts of climate change.


Subject(s)
Climate Change , Communicable Diseases , Animals , United States/epidemiology , Communicable Diseases/epidemiology , Public Health , Weather , Temperature
5.
Clin Infect Dis ; 76(5): 850-860, 2023 03 04.
Article in English | MEDLINE | ID: mdl-36268576

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) reinfection is poorly understood, partly because few studies have systematically applied genomic analysis to distinguish reinfection from persistent RNA detection related to initial infection. We aimed to evaluate the characteristics of SARS-CoV-2 reinfection and persistent RNA detection using independent genomic, clinical, and laboratory assessments. METHODS: All individuals at a large academic medical center who underwent a SARS-CoV-2 nucleic acid amplification test (NAAT) ≥45 days after an initial positive test, with both tests between 14 March and 30 December 2020, were analyzed for potential reinfection. Inclusion criteria required having ≥2 positive NAATs collected ≥45 days apart with a cycle threshold (Ct) value <35 at repeat testing. For each included subject, likelihood of reinfection was assessed by viral genomic analysis of all available specimens with a Ct value <35, structured Ct trajectory criteria, and case-by-case review by infectious diseases physicians. RESULTS: Among 1569 individuals with repeat SARS-CoV-2 testing ≥45 days after an initial positive NAAT, 65 (4%) met cohort inclusion criteria. Viral genomic analysis characterized mutations present and was successful for 14/65 (22%) subjects. Six subjects had genomically supported reinfection, and 8 subjects had genomically supported persistent RNA detection. Compared to viral genomic analysis, clinical and laboratory assessments correctly distinguished reinfection from persistent RNA detection in 12/14 (86%) subjects but missed 2/6 (33%) genomically supported reinfections. CONCLUSIONS: Despite good overall concordance with viral genomic analysis, clinical and Ct value-based assessments failed to identify 33% of genomically supported reinfections. Scaling-up genomic analysis for clinical use would improve detection of SARS-CoV-2 reinfections.


Subject(s)
COVID-19 , Humans , COVID-19/diagnosis , COVID-19 Testing , Reinfection/diagnosis , Retrospective Studies , SARS-CoV-2/genetics , RNA
6.
J Clin Microbiol ; 61(5): e0132622, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37022168

ABSTRACT

A bacterial species is considered to be intrinsically resistant to an antimicrobial when nearly all of the wild-type isolates (i.e., those without acquired resistance) exhibit minimum inhibitory concentration (MIC) values that are sufficiently high such that susceptibility testing is unnecessary, and that the antimicrobial should not be considered for therapy. Accordingly, knowledge of intrinsic resistance influences both the selection of treatment regimens and the approach to susceptibility testing in the clinical laboratory, where unexpected results also facilitate the recognition of microbial identification or susceptibility testing errors. Previously, limited data have suggested that Hafnia spp. may be intrinsically resistant to colistin. We evaluated the in vitro activity of colistin against 119 Hafniaceae that were isolated from human samples: 75 (63%) from routine clinical cultures and 44 (37%) from stool samples of travelers undergoing screening for antimicrobial resistant organisms. Broth microdilution colistin MICs were ≥4 µg/mL for 117 of 119 (98%) isolates. Whole-genome sequencing of 96 of the isolates demonstrated that the colistin-resistant phenotype was not lineage-specific. 2 of the 96 (2%) isolates harbored mobile colistin resistance genes. Compared to whole-genome sequencing, VITEK MS matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and VITEK 2 GN ID failed to consistently distinguish between Hafnia alvei, Hafnia paralvei, and Obesumbacterium proteus. In conclusion, using a reference antimicrobial susceptibility testing method and a genetically diverse collection of isolates, we found Hafnia spp. to be intrinsically resistant to colistin. The recognition of this phenotype will help inform rational approaches by which to perform antimicrobial susceptibility testing and therapy for patients with infections that are caused by Hafnia spp.


Subject(s)
Anti-Infective Agents , Hafnia , Humans , Colistin/pharmacology , Enterobacteriaceae , Hafnia/genetics , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology
7.
Cardiovasc Diabetol ; 21(1): 136, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35864532

ABSTRACT

BACKGROUND: The high heterogeneity in the symptoms and severity of COVID-19 makes it challenging to identify high-risk patients early in the disease. Cardiometabolic comorbidities have shown strong associations with COVID-19 severity in epidemiologic studies. Cardiometabolic protein biomarkers, therefore, may provide predictive insight regarding which patients are most susceptible to severe illness from COVID-19. METHODS: In plasma samples collected from 343 patients hospitalized with COVID-19 during the first wave of the pandemic, we measured 92 circulating protein biomarkers previously implicated in cardiometabolic disease. We performed proteomic analysis and developed predictive models for severe outcomes. We then used these models to predict the outcomes of out-of-sample patients hospitalized with COVID-19 later in the surge (N = 194). RESULTS: We identified a set of seven protein biomarkers predictive of admission to the intensive care unit and/or death (ICU/death) within 28 days of presentation to care. Two of the biomarkers, ADAMTS13 and VEGFD, were associated with a lower risk of ICU/death. The remaining biomarkers, ACE2, IL-1RA, IL6, KIM1, and CTSL1, were associated with higher risk. When used to predict the outcomes of the future, out-of-sample patients, the predictive models built with these protein biomarkers outperformed all models built from standard clinical data, including known COVID-19 risk factors. CONCLUSIONS: These findings suggest that proteomic profiling can inform the early clinical impression of a patient's likelihood of developing severe COVID-19 outcomes and, ultimately, accelerate the recognition and treatment of high-risk patients.


Subject(s)
COVID-19 , Cardiovascular Diseases , Biomarkers , Cardiovascular Diseases/diagnosis , Humans , Proteomics , SARS-CoV-2
8.
Am J Public Health ; 112(11): 1556-1559, 2022 11.
Article in English | MEDLINE | ID: mdl-36223583

ABSTRACT

Mobile health units can improve access to preventive health services, especially for medically underserved populations. However, there is little published experience of mobile health units being used to expand access to COVID-19 vaccination. In concert with local public health departments and community members, we implemented a mobile COVID-19 health unit and deployed it to 12 predominantly low-income and racial/ethnic minority communities in Massachusetts. We describe the success and challenges of this innovative program in expanding access to COVID-19 vaccination. (Am J Public Health. 2022;112(11):1556-1559. https://doi.org/10.2105/AJPH.2022.307021).


Subject(s)
COVID-19 , Medically Underserved Area , COVID-19/prevention & control , COVID-19 Vaccines , Counseling , Ethnicity , Health Services Accessibility , Humans , Minority Groups , Vaccination
9.
Prev Med ; 163: 107226, 2022 10.
Article in English | MEDLINE | ID: mdl-36029925

ABSTRACT

COVID-19 has disproportionately impacted underserved populations, including racial/ethnic minorities. Prior studies have demonstrated that mobile health units are effective at expanding preventive services for hard-to-reach populations, but this has not been studied in the context of COVID-19 vaccination. Our objective was to determine if voluntary participants who access mobile COVID-19 vaccination units are more likely to be racial/ethnic minorities and adolescents compared with the general vaccinated population. We conducted a cross-sectional study of individuals who presented to three different mobile COVID-19 vaccination units in the Greater Boston area from May 20, 2021, to August 18, 2021. We acquired data regarding the general vaccinated population in the state and of target communities from the Massachusetts Department of Public Health. We used chi-square testing to compare the demographic characteristics of mobile vaccination unit participants and the general state and community populations that received COVID-19 vaccines during the same time period. We found that during this three-month period, mobile vaccination units held 130 sessions and administered 2622 COVID-19 vaccine doses to 1982 unique participants. The median (IQR) age of participants was 31 (16-46) years, 1016 (51%) were female, 1575 (80%) were non-White, and 1126 (57%) were Hispanic. Participants in the mobile vaccination units were more likely to be younger (p < 0.001), non-White race (p < 0.001), and Hispanic ethnicity (p < 0.001) compared with the general vaccinated population of the state and target communities. This study suggests that mobile vaccination units have the potential to improve access to COVID-19 vaccination for diverse populations.


Subject(s)
COVID-19 Vaccines , COVID-19 , Adolescent , Adult , COVID-19/epidemiology , COVID-19/prevention & control , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Mobile Health Units , Vaccination , Vulnerable Populations
10.
Teach Learn Med ; 34(3): 329-340, 2022.
Article in English | MEDLINE | ID: mdl-34011226

ABSTRACT

Issue: As U.S. healthcare systems plan for future physician workforce needs, the systemic impacts of climate change, a worldwide environmental and health crisis, have not been factored in. The current focus on increasing the number of trained physicians and optimizing efficiencies in healthcare delivery may be insufficient. Graduate medical education (GME) priorities and training should be considered in order to prepare a climate-educated physician workforce. Evidence: We used a holistic lens to explore the available literature regarding the intersection of future physician workforce needs, GME program priorities, and resident education within the larger context of climate change. Our interinstitutional, transdisciplinary team brought perspectives from their own fields, including climate science, climate and health research, and medical education to provide recommendations for building a climate-educated physician workforce. Implications: Acknowledging and preparing for the effects of climate change on the physician workforce will require identification of workforce gaps, changes to GME program priorities, and education of trainees on the health and societal impacts of climate change. Alignment of GME training with workforce considerations and climate action and adaptation initiatives will be critical in ensuring the U.S. has a climate-educated physician workforce capable of addressing health and healthcare system challenges. This article offers a number of recommendations for physician workforce priorities, resident education, and system-level changes to better prepare for the health and health system impacts of climate change.


Subject(s)
Internship and Residency , Medicine , Physicians , Climate Change , Education, Medical, Graduate , Humans , United States , Workforce
11.
J Infect Dis ; 223(2): 342-351, 2021 02 03.
Article in English | MEDLINE | ID: mdl-32610345

ABSTRACT

BACKGROUND: Susceptibility to Vibrio cholerae infection is affected by blood group, age, and preexisting immunity, but these factors only partially explain who becomes infected. A recent study used 16S ribosomal RNA amplicon sequencing to quantify the composition of the gut microbiome and identify predictive biomarkers of infection with limited taxonomic resolution. METHODS: To achieve increased resolution of gut microbial factors associated with V. cholerae susceptibility and identify predictors of symptomatic disease, we applied deep shotgun metagenomic sequencing to a cohort of household contacts of patients with cholera. RESULTS: Using machine learning, we resolved species, strains, gene families, and cellular pathways in the microbiome at the time of exposure to V. cholerae to identify markers that predict infection and symptoms. Use of metagenomic features improved the precision and accuracy of prediction relative to 16S sequencing. We also predicted disease severity, although with greater uncertainty than our infection prediction. Species within the genera Prevotella and Bifidobacterium predicted protection from infection, and genes involved in iron metabolism were also correlated with protection. CONCLUSION: Our results highlight the power of metagenomics to predict disease outcomes and suggest specific species and genes for experimental testing to investigate mechanisms of microbiome-related protection from cholera.


Subject(s)
Cholera/diagnosis , Cholera/microbiology , Metagenomics , Vibrio cholerae/physiology , Biomarkers , Disease Susceptibility , Gastrointestinal Microbiome , Metagenome , Metagenomics/methods , Phylogeny , Prognosis , ROC Curve , Severity of Illness Index
12.
Infect Immun ; 89(9): e0021721, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34228490

ABSTRACT

Cholera is a diarrheal disease caused by Vibrio cholerae that continues to be a major public health concern in populations without access to safe water. IgG- and IgA-secreting memory B cells (MBC) targeting the V. cholerae O-specific polysaccharide (OSP) correlate with protection from infection in persons exposed to V. cholerae and may be a major determinant of long-term protection against cholera. Shanchol, a widely used oral cholera vaccine (OCV), stimulates OSP MBC responses in only some people after vaccination, and the gut microbiota is a possible determinant of variable immune responses observed after OCV. Using 16S rRNA sequencing of feces from the time of vaccination, we compared the gut microbiota among adults with and without MBC responses to OCV. Gut microbial diversity measures were not associated with MBC isotype or OSP-specific responses, but individuals with a higher abundance of Clostridiales and lower abundance of Enterobacterales were more likely to develop an MBC response. We applied protein-normalized fecal supernatants of high and low MBC responders to THP-1-derived human macrophages to investigate the effect of microbial factors at the time of vaccination. Feces from individuals with higher MBC responses induced significantly different IL-1ß and IL-6 levels than individuals with lower responses, indicating that the gut microbiota at the time of vaccination may "prime" the mucosal immune response to vaccine antigens. Our results suggest the gut microbiota could impact immune responses to OCVs, and further study of microbial metabolites as potential vaccine adjuvants is warranted.


Subject(s)
B-Lymphocytes/immunology , Cholera Vaccines/immunology , Cholera/immunology , Cholera/microbiology , Gastrointestinal Microbiome , Immunologic Memory , Vibrio cholerae/immunology , Administration, Oral , Adolescent , Adult , Antibody Specificity/immunology , B-Lymphocytes/metabolism , Cholera/prevention & control , Cholera Vaccines/administration & dosage , Female , Host-Pathogen Interactions/immunology , Humans , Male , Microbial Interactions , Vaccination , Young Adult
13.
Curr Opin Infect Dis ; 34(5): 423-431, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34267046

ABSTRACT

PURPOSE OF REVIEW: Antimicrobial resistance (AMR) in bacteria poses a major risk to global public health, with many factors contributing to the observed increase in AMR. International travel is one recognized contributor. The purpose of this review is to summarize current knowledge regarding the acquisition, carriage and spread of AMR bacteria by international travelers. RECENT FINDINGS: Recent studies have highlighted that travel is an important risk factor for the acquisition of AMR bacteria, with approximately 30% of studied travelers returning with an acquired AMR bacterium. Epidemiological studies have shown there are three major risk factors for acquisition: travel destination, antimicrobial usage and travelers' diarrhea (TD). Analyses have begun to illustrate the AMR genes that are acquired and spread by travelers, risk factors for acquisition and carriage of AMR bacteria, and local transmission of imported AMR organisms. SUMMARY: International travel is a contributor to the acquisition and dissemination of AMR organisms globally. Efforts to reduce the burden of AMR organisms should include a focus on international travelers. Routine genomic surveillance would further elucidate the role of international travel in the global spread of AMR bacteria.


Subject(s)
Diarrhea , Travel , Anti-Bacterial Agents/therapeutic use , Bacteria , Diarrhea/drug therapy , Global Health , Humans
14.
Clin Infect Dis ; 69(2): 306-315, 2019 07 02.
Article in English | MEDLINE | ID: mdl-30312374

ABSTRACT

BACKGROUND: Measles importations and the subsequent spread from US travelers returning from abroad are responsible for most measles cases in the United States. Increasing measles-mumps-rubella (MMR) vaccination among departing US travelers could reduce the clinical impact and costs of measles in the United States. METHODS: We designed a decision tree to evaluate MMR vaccination at a pretravel health encounter (PHE), compared with no encounter. We derived input parameters from Global TravEpiNet data and literature. We quantified Riskexposure to measles while traveling and the average number of US-acquired cases and contacts due to a measles importation. In sensitivity analyses, we examined the impact of destination-specific Riskexposure, including hot spots with active measles outbreaks; the percentage of previously-unvaccinated travelers; and the percentage of travelers returning to US communities with heterogeneous MMR coverage. RESULTS: The no-encounter strategy projected 22 imported and 66 US-acquired measles cases, costing $14.8M per 10M travelers. The PHE strategy projected 15 imported and 35 US-acquired cases at $190.3M per 10M travelers. PHE was not cost effective for all international travelers (incremental cost-effectiveness ratio [ICER] $4.6M/measles case averted), but offered better value (ICER <$100 000/measles case averted) or was even cost saving for travelers to hot spots, especially if travelers were previously unvaccinated or returning to US communities with heterogeneous MMR coverage. CONCLUSIONS: PHEs that improve MMR vaccination among US international travelers could reduce measles cases, but are costly. The best value is for travelers with a high likelihood of measles exposure, especially if the travelers are previously unvaccinated or will return to US communities with heterogeneous MMR coverage.


Subject(s)
Communicable Diseases, Imported/economics , Communicable Diseases, Imported/prevention & control , Cost-Benefit Analysis , Measles-Mumps-Rubella Vaccine/economics , Measles/economics , Measles/prevention & control , Travel-Related Illness , Adult , Communicable Diseases, Imported/epidemiology , Female , Humans , Male , Measles/epidemiology , Measles-Mumps-Rubella Vaccine/administration & dosage , Treatment Outcome , United States/epidemiology , Young Adult
15.
Ann Intern Med ; 168(1): ITC1-ITC16, 2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29297035

ABSTRACT

International travel can result in new illness or exacerbate existing conditions, and primary care clinicians have the opportunity to provide both pre- and posttravel health care. Providers should be familiar with destination-specific disease risks, be knowledgeable about travel and routine vaccines, be prepared to prescribe chemoprophylaxis and self-treatment regimens, and be aware of travel medicine resources.


Subject(s)
Primary Health Care , Travel Medicine , Accidents, Traffic/prevention & control , Aircraft , Altitude Sickness/prevention & control , Animals , Barotrauma/prevention & control , Food Safety , Humans , Insect Bites and Stings/prevention & control , Sexually Transmitted Diseases/prevention & control , Ships , United States , Wounds and Injuries/prevention & control , Zoonoses/prevention & control
16.
Ann Intern Med ; 176(12): eL230347, 2023 12.
Article in English | MEDLINE | ID: mdl-38109751
17.
JAMA ; 331(15): 1318-1319, 2024 04 16.
Article in English | MEDLINE | ID: mdl-38506835

ABSTRACT

This JAMA Insights in the Climate Change and Health series discusses the importance of clinicians having awareness of changes in the geographic range, seasonality, and intensity of transmission of infectious diseases to help them diagnose, treat, and prevent these diseases.


Subject(s)
Climate Change , Communicable Diseases , Humans , Communicable Diseases/diagnosis , Communicable Diseases/epidemiology , Climatic Processes , Extreme Weather , Wildfires , Greenhouse Gases/adverse effects , Fossil Fuels/adverse effects , Disease Vectors , Zoonoses/epidemiology , Mycoses/epidemiology , Waterborne Diseases/epidemiology , Education, Medical , Public Policy
18.
J Infect Dis ; 218(4): 645-653, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29659916

ABSTRACT

Background: Cholera is a public health problem worldwide, and the risk factors for infection are only partially understood. Methods: We prospectively studied household contacts of patients with cholera to compare those who were infected to those who were not. We constructed predictive machine learning models of susceptibility, using baseline gut microbiota data. We identified bacterial taxa associated with susceptibility to Vibrio cholerae infection and tested these taxa for interactions with V. cholerae in vitro. Results: We found that machine learning models based on gut microbiota, as well as models based on known clinical and epidemiological risk factors, predicted V. cholerae infection. A predictive gut microbiota of roughly 100 bacterial taxa discriminated between contacts who developed infection and those who did not. Susceptibility to cholera was associated with depleted levels of microbes from the phylum Bacteroidetes. By contrast, a microbe associated with cholera by our modeling framework, Paracoccus aminovorans, promoted the in vitro growth of V. cholerae. Gut microbiota structure, clinical outcome, and age were also linked. Conclusion: These findings support the hypothesis that abnormal gut microbial communities are a host factor related to V. cholerae susceptibility.


Subject(s)
Cholera/epidemiology , Cholera/immunology , Disease Susceptibility , Gastrointestinal Microbiome , Microbiota , Vibrio cholerae/growth & development , Vibrio cholerae/immunology , Adolescent , Adult , Child , Child, Preschool , Computer Simulation , Epidemiologic Methods , Family Characteristics , Family Health , Female , Humans , Machine Learning , Male , Middle Aged , Prospective Studies , Risk Factors , Young Adult
19.
Infect Immun ; 86(2)2018 02.
Article in English | MEDLINE | ID: mdl-29133347

ABSTRACT

To better understand the innate immune response to Vibrio cholerae infection, we tracked gene expression in the duodenal mucosa of 11 Bangladeshi adults with cholera, using biopsy specimens obtained immediately after rehydration and 30 and 180 days later. We identified differentially expressed genes and performed an analysis to predict differentially regulated pathways and upstream regulators. During acute cholera, there was a broad increase in the expression of genes associated with innate immunity, including activation of the NF-κB, mitogen-activated protein kinase (MAPK), and Toll-like receptor (TLR)-mediated signaling pathways, which, unexpectedly, persisted even 30 days after infection. Focusing on early differences in gene expression, we identified 37 genes that were differentially expressed on days 2 and 30 across the 11 participants. These genes included the endosomal Toll-like receptor gene TLR8, which was expressed in lamina propria cells. Underscoring a potential role for endosomal TLR-mediated signaling in vivo, our pathway analysis found that interferon regulatory factor 7 and beta 1 and alpha 2 interferons were among the top upstream regulators activated during cholera. Among the innate immune effectors, we found that the gene for DUOX2, an NADPH oxidase involved in the maintenance of intestinal homeostasis, was upregulated in intestinal epithelial cells during cholera. Notably, the observed increases in DUOX2 and TLR8 expression were also modeled in vitro when Caco-2 or THP-1 cells, respectively, were stimulated with live V. cholerae but not with heat-killed organisms or cholera toxin alone. These previously unidentified features of the innate immune response to V. cholerae extend our understanding of the mucosal immune signaling pathways and effectors activated in vivo following cholera.


Subject(s)
Cholera/immunology , Immunity, Innate , Immunity, Mucosal , Signal Transduction , Vibrio cholerae/immunology , Adult , Biopsy , Cholera/pathology , Duodenum/pathology , Female , Gene Expression Profiling , Humans , Male , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL