Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Clin Microbiol ; 61(4): e0163422, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37010411

ABSTRACT

Offering patients with tuberculosis (TB) an optimal and timely treatment regimen depends on the rapid detection of Mycobacterium tuberculosis (Mtb) drug resistance from clinical samples. Finding Low Abundance Sequences by Hybridization (FLASH) is a technique that harnesses the efficiency, specificity, and flexibility of the Cas9 enzyme to enrich targeted sequences. Here, we used FLASH to amplify 52 candidate genes probably associated with resistance to first- and second-line drugs in the Mtb reference strain (H37Rv), then detect drug resistance mutations in cultured Mtb isolates, and in sputum samples. 92% of H37Rv reads mapped to Mtb targets, with 97.8% of target regions covered at a depth ≥ 10X. Among cultured isolates, FLASH-TB detected the same 17 drug resistance mutations as whole genome sequencing (WGS) did, but with much greater depth. Among the 16 sputum samples, FLASH-TB increased recovery of Mtb DNA compared with WGS (from 1.4% [IQR 0.5-7.5] to 33% [IQR 4.6-66.3]) and average depth reads of targets (from 6.3 [IQR 3.8-10.5] to 1991 [IQR 254.4-3623.7]). FLASH-TB identified Mtb complex in all 16 samples based on IS1081 and IS6110 copies. Drug resistance predictions for 15/16 (93.7%) clinical samples were highly concordant with phenotypic DST for isoniazid, rifampicin, amikacin, and kanamycin [15/15 (100%)], ethambutol [12/15 (80%)] and moxifloxacin [14/15 (93.3%)]. These results highlighted the potential of FLASH-TB for detecting Mtb drug resistance from sputum samples.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis, Multidrug-Resistant , Tuberculosis , Humans , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Sputum/microbiology , Tuberculosis, Multidrug-Resistant/microbiology , Tuberculosis/drug therapy , Mycobacterium tuberculosis/genetics , Microbial Sensitivity Tests
2.
J Clin Invest ; 132(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35113809

ABSTRACT

BackgroundBacterial vaginosis (BV) causes genital inflammation and increases HIV risk, whereas a vaginal microbiota dominated by Lactobacillus species is associated with immune quiescence and relative HIV protection. BV treatment reduces genital inflammation, but it is unclear whether this reduction is driven by a decrease in BV-associated bacteria or an increase in Lactobacillus species.METHODSTo evaluate the short-term effect of standard BV treatment on genital immunology and the vaginal microbiota, vaginal swabs were collected immediately before and after metronidazole treatment for BV and analyzed with multiplex ELISA, metagenomic sequencing, and quantitative PCR.RESULTSTopical metronidazole treatment rapidly reduced vaginal levels of proinflammatory cytokines, chemokines, and soluble immune markers of epithelial barrier disruption. Although the vaginal microbiota shifted to dominance by L. iners or L. jensenii, this proportional shift was primarily driven by a 2 to 4 log10-fold reduction in BV-associated bacteria absolute abundance. BV treatment induced no change in the absolute abundance of L. crispatus or L. iners and only minor (<1 log10-fold) increases in L. gasseri and L. jensenii that were not independently associated with reduced inflammation in multivariable models.CONCLUSIONThe genital immune benefits that are associated with Lactobacillus dominance after BV treatment were not directly attributable to an absolute increase in lactobacilli, but rather to the loss of BV-associated bacteria.Trial REGISTRATIONParticipants were recruited as part of a randomized controlled trial (ClinicalTrials.gov NCT02766023) from 2016 to 2019.FUNDINGCanadian Institutes of Health Research (PJT-156123) and the National Institute of Allergy and Infectious Diseases (HHSN2722013000141 and HHSN27200007).


Subject(s)
HIV Infections , Vaginosis, Bacterial , Bacteria , Female , HIV Infections/prevention & control , Humans , Inflammation/drug therapy , Lactobacillus , Metronidazole/pharmacology , Vagina
4.
Cell ; 184(2): 323-333.e9, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33306959

ABSTRACT

The December 2019 outbreak of a novel respiratory virus, SARS-CoV-2, has become an ongoing global pandemic due in part to the challenge of identifying symptomatic, asymptomatic, and pre-symptomatic carriers of the virus. CRISPR diagnostics can augment gold-standard PCR-based testing if they can be made rapid, portable, and accurate. Here, we report the development of an amplification-free CRISPR-Cas13a assay for direct detection of SARS-CoV-2 from nasal swab RNA that can be read with a mobile phone microscope. The assay achieved ∼100 copies/µL sensitivity in under 30 min of measurement time and accurately detected pre-extracted RNA from a set of positive clinical samples in under 5 min. We combined crRNAs targeting SARS-CoV-2 RNA to improve sensitivity and specificity and directly quantified viral load using enzyme kinetics. Integrated with a reader device based on a mobile phone, this assay has the potential to enable rapid, low-cost, point-of-care screening for SARS-CoV-2.


Subject(s)
COVID-19 Nucleic Acid Testing/methods , Cell Phone/instrumentation , Optical Imaging/methods , RNA, Viral/analysis , Viral Load/methods , Animals , COVID-19 Nucleic Acid Testing/economics , COVID-19 Nucleic Acid Testing/instrumentation , CRISPR-Cas Systems , Cell Line , Coronavirus Nucleocapsid Proteins/genetics , Humans , Nasopharynx/virology , Optical Imaging/instrumentation , Phosphoproteins/genetics , Point-of-Care Testing , RNA Interference , RNA, Viral/genetics , Sensitivity and Specificity , Viral Load/economics , Viral Load/instrumentation
5.
Nat Methods ; 17(12): 1183-1190, 2020 12.
Article in English | MEDLINE | ID: mdl-33077967

ABSTRACT

CRISPR-Cas technologies have enabled programmable gene editing in eukaryotes and prokaryotes. However, the leading Cas9 and Cas12a enzymes are limited in their ability to make large deletions. Here, we used the processive nuclease Cas3, together with a minimal Type I-C Cascade-based system for targeted genome engineering in bacteria. DNA cleavage guided by a single CRISPR RNA generated large deletions (7-424 kilobases) in Pseudomonas aeruginosa with near-100% efficiency, while Cas9 yielded small deletions and point mutations. Cas3 generated bidirectional deletions originating from the programmed site, which was exploited to reduce the P. aeruginosa genome by 837 kb (13.5%). Large deletion boundaries were efficiently specified by a homology-directed repair template during editing with Cascade-Cas3, but not Cas9. A transferable 'all-in-one' vector was functional in Escherichia coli, Pseudomonas syringae and Klebsiella pneumoniae, and endogenous CRISPR-Cas use was enhanced with an 'anti-anti-CRISPR' strategy. P. aeruginosa Type I-C Cascade-Cas3 (PaeCas3c) facilitates rapid strain manipulation with applications in synthetic biology, genome minimization and the removal of large genomic regions.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/genetics , DNA Helicases/metabolism , Escherichia coli Proteins/metabolism , Gene Editing/methods , Genetic Engineering/methods , Base Sequence/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Escherichia coli/genetics , Genome, Bacterial/genetics , Klebsiella pneumoniae/genetics , Pseudomonas aeruginosa/genetics , Pseudomonas syringae/genetics , Sequence Deletion/genetics
8.
Article in English | MEDLINE | ID: mdl-32139440

ABSTRACT

OBJECTIVE: In 2016, Catalonia experienced a pediatric brainstem encephalitis outbreak caused by enterovirus A71 (EV-A71). Conventional testing identified EV in the periphery but rarely in CSF. Metagenomic next-generation sequencing (mNGS) and CSF pan-viral serology (VirScan) were deployed to enhance viral detection and characterization. METHODS: RNA was extracted from the CSF (n = 20), plasma (n = 9), stool (n = 15), and nasopharyngeal samples (n = 16) from 10 children with brainstem encephalitis and 10 children with meningitis or encephalitis. Pathogens were identified using mNGS. Available CSF from cases (n = 12) and pediatric other neurologic disease controls (n = 54) were analyzed with VirScan with a subset (n = 9 and n = 50) validated by ELISA. RESULTS: mNGS detected EV in all samples positive by quantitative reverse transcription polymerase chain reaction (qRT-PCR) (n = 25). In qRT-PCR-negative samples (n = 35), mNGS found virus in 23% (n = 8, 3 CSF samples). Overall, mNGS enhanced EV detection from 42% (25/60) to 57% (33/60) (p-value = 0.013). VirScan and ELISA increased detection to 92% (11/12) compared with 46% (4/12) for CSF mNGS and qRT-PCR (p-value = 0.023). Phylogenetic analysis confirmed the EV-A71 strain clustered with a neurovirulent German EV-A71. A single amino acid substitution (S241P) in the EVA71 VP1 protein was exclusive to the CNS in one subject. CONCLUSION: mNGS with VirScan significantly increased the CNS detection of EVs relative to qRT-PCR, and the latter generated an antigenic profile of the acute EV-A71 immune response. Genomic analysis confirmed the close relation of the outbreak EV-A71 and neuroinvasive German EV-A71. A S241P substitution in VP1 was found exclusively in the CSF.


Subject(s)
Brain Stem , Encephalitis, Viral/virology , Enterovirus A, Human/genetics , Enterovirus A, Human/isolation & purification , Enterovirus Infections/virology , Meningitis, Viral/virology , RNA, Viral/metabolism , Child, Preschool , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Phylogeny , RNA, Viral/blood , RNA, Viral/cerebrospinal fluid , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA
9.
mBio ; 10(6)2019 12 17.
Article in English | MEDLINE | ID: mdl-31848287

ABSTRACT

The burden of meningitis in low-and-middle-income countries remains significant, but the infectious causes remain largely unknown, impeding institution of evidence-based treatment and prevention decisions. We conducted a validation and application study of unbiased metagenomic next-generation sequencing (mNGS) to elucidate etiologies of meningitis in Bangladesh. This RNA mNGS study was performed on cerebrospinal fluid (CSF) specimens from patients admitted in the largest pediatric hospital, a World Health Organization sentinel site, with known neurologic infections (n = 36), with idiopathic meningitis (n = 25), and with no infection (n = 30), and six environmental samples, collected between 2012 and 2018. We used the IDseq bioinformatics pipeline and machine learning to identify potentially pathogenic microbes, which we then confirmed orthogonally and followed up through phone/home visits. In samples with known etiology and without infections, there was 83% concordance between mNGS and conventional testing. In idiopathic cases, mNGS identified a potential bacterial or viral etiology in 40%. There were three instances of neuroinvasive Chikungunya virus (CHIKV), whose genomes were >99% identical to each other and to a Bangladeshi strain only previously recognized to cause febrile illness in 2017. CHIKV-specific qPCR of all remaining stored CSF samples from children who presented with idiopathic meningitis in 2017 (n = 472) revealed 17 additional CHIKV meningitis cases, exposing an unrecognized meningitis outbreak. Orthogonal molecular confirmation, case-based clinical data, and patient follow-up substantiated the findings. Case-control CSF mNGS surveys can complement conventional diagnostic methods to identify etiologies of meningitis, conduct surveillance, and predict outbreaks. The improved patient- and population-level data can inform evidence-based policy decisions.IMPORTANCE Globally, there are an estimated 10.6 million cases of meningitis and 288,000 deaths every year, with the vast majority occurring in low- and middle-income countries. In addition, many survivors suffer from long-term neurological sequelae. Most laboratories assay only for common bacterial etiologies using culture and directed PCR, and the majority of meningitis cases lack microbiological diagnoses, impeding institution of evidence-based treatment and prevention strategies. We report here the results of a validation and application study of using unbiased metagenomic sequencing to determine etiologies of idiopathic (of unknown cause) cases. This included CSF from patients with known neurologic infections, with idiopathic meningitis, and without infection admitted in the largest children's hospital of Bangladesh and environmental samples. Using mNGS and machine learning, we identified and confirmed an etiology (viral or bacterial) in 40% of idiopathic cases. We detected three instances of Chikungunya virus (CHIKV) that were >99% identical to each other and to a strain previously recognized to cause systemic illness only in 2017. CHIKV qPCR of all remaining stored 472 CSF samples from children who presented with idiopathic meningitis in 2017 at the same hospital uncovered an unrecognized CHIKV meningitis outbreak. CSF mNGS can complement conventional diagnostic methods to identify etiologies of meningitis, and the improved patient- and population-level data can inform better policy decisions.


Subject(s)
Chikungunya virus/genetics , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/virology , Disease Outbreaks , Genome, Viral , Meningitis, Viral/epidemiology , Meningitis, Viral/virology , Metagenomics , Bangladesh/epidemiology , Chikungunya virus/classification , Chikungunya virus/immunology , Female , Humans , Infant , Infant, Newborn , Male , Meningitis, Viral/diagnosis , Meningitis, Viral/immunology , Metagenomics/methods , Phylogeny , Public Health Surveillance
10.
Nat Med ; 25(11): 1748-1752, 2019 11.
Article in English | MEDLINE | ID: mdl-31636453

ABSTRACT

Since 2012, the United States of America has experienced a biennial spike in pediatric acute flaccid myelitis (AFM)1-6. Epidemiologic evidence suggests non-polio enteroviruses (EVs) are a potential etiology, yet EV RNA is rarely detected in cerebrospinal fluid (CSF)2. CSF from children with AFM (n = 42) and other pediatric neurologic disease controls (n = 58) were investigated for intrathecal antiviral antibodies, using a phage display library expressing 481,966 overlapping peptides derived from all known vertebrate and arboviruses (VirScan). Metagenomic next-generation sequencing (mNGS) of AFM CSF RNA (n = 20 cases) was also performed, both unbiased sequencing and with targeted enrichment for EVs. Using VirScan, the viral family significantly enriched by the CSF of AFM cases relative to controls was Picornaviridae, with the most enriched Picornaviridae peptides belonging to the genus Enterovirus (n = 29/42 cases versus 4/58 controls). EV VP1 ELISA confirmed this finding (n = 22/26 cases versus 7/50 controls). mNGS did not detect additional EV RNA. Despite rare detection of EV RNA, pan-viral serology frequently identified high levels of CSF EV-specific antibodies in AFM compared with controls, providing further evidence for a causal role of non-polio EVs in AFM.


Subject(s)
Central Nervous System Viral Diseases/genetics , Enterovirus Infections/genetics , Enterovirus/genetics , Myelitis/genetics , Neuromuscular Diseases/genetics , Seroepidemiologic Studies , Antibodies, Viral/cerebrospinal fluid , Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Central Nervous System Viral Diseases/cerebrospinal fluid , Central Nervous System Viral Diseases/epidemiology , Central Nervous System Viral Diseases/virology , Child, Preschool , Enterovirus/pathogenicity , Enterovirus Infections/cerebrospinal fluid , Enterovirus Infections/epidemiology , Enterovirus Infections/virology , Female , Humans , Infant , Male , Myelitis/cerebrospinal fluid , Myelitis/epidemiology , Myelitis/virology , Neuromuscular Diseases/cerebrospinal fluid , Neuromuscular Diseases/epidemiology , Neuromuscular Diseases/virology , United States
12.
Nucleic Acids Res ; 47(14): e83, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31114866

ABSTRACT

The growing prevalence of deadly microbes with resistance to previously life-saving drug therapies is a dire threat to human health. Detection of low abundance pathogen sequences remains a challenge for metagenomic Next Generation Sequencing (NGS). We introduce FLASH (Finding Low Abundance Sequences by Hybridization), a next-generation CRISPR/Cas9 diagnostic method that takes advantage of the efficiency, specificity and flexibility of Cas9 to enrich for a programmed set of sequences. FLASH-NGS achieves up to 5 orders of magnitude of enrichment and sub-attomolar gene detection with minimal background. We provide an open-source software tool (FLASHit) for guide RNA design. Here we applied it to detection of antimicrobial resistance genes in respiratory fluid and dried blood spots, but FLASH-NGS is applicable to all areas that rely on multiplex PCR.


Subject(s)
Anti-Bacterial Agents/pharmacology , CRISPR-Cas Systems , Computational Biology/methods , Drug Resistance, Bacterial/drug effects , High-Throughput Nucleotide Sequencing/methods , Bacteria/classification , Bacteria/drug effects , Bacteria/genetics , Bacterial Infections/diagnosis , Bacterial Infections/genetics , Bacterial Infections/prevention & control , Drug Resistance, Bacterial/genetics , Humans , Metagenomics/methods , Reproducibility of Results , Sensitivity and Specificity
14.
Clin Infect Dis ; 68(11): 1847-1855, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30239621

ABSTRACT

BACKGROUND: Despite improved diagnostics, pulmonary pathogens in immunocompromised children frequently evade detection, leading to significant mortality. Therefore, we aimed to develop a highly sensitive metagenomic next-generation sequencing (mNGS) assay capable of evaluating the pulmonary microbiome and identifying diverse pathogens in the lungs of immunocompromised children. METHODS: We collected 41 lower respiratory specimens from 34 immunocompromised children undergoing evaluation for pulmonary disease at 3 children's hospitals from 2014-2016. Samples underwent mechanical homogenization, parallel RNA/DNA extraction, and metagenomic sequencing. Sequencing reads were aligned to the National Center for Biotechnology Information nucleotide reference database to determine taxonomic identities. Statistical outliers were determined based on abundance within each sample and relative to other samples in the cohort. RESULTS: We identified a rich cross-domain pulmonary microbiome that contained bacteria, fungi, RNA viruses, and DNA viruses in each patient. Potentially pathogenic bacteria were ubiquitous among samples but could be distinguished as possible causes of disease by parsing for outlier organisms. Samples with bacterial outliers had significantly depressed alpha-diversity (median, 0.61; interquartile range [IQR], 0.33-0.72 vs median, 0.96; IQR, 0.94-0.96; P < .001). Potential pathogens were detected in half of samples previously negative by clinical diagnostics, demonstrating increased sensitivity for missed pulmonary pathogens (P < .001). CONCLUSIONS: An optimized mNGS assay for pulmonary microbes demonstrates significant inoculation of the lower airways of immunocompromised children with diverse bacteria, fungi, and viruses. Potential pathogens can be identified based on absolute and relative abundance. Ongoing investigation is needed to determine the pathogenic significance of outlier microbes in the lungs of immunocompromised children with pulmonary disease.


Subject(s)
Immunocompromised Host , Lung Diseases/microbiology , Lung Diseases/virology , Lung/microbiology , Lung/virology , Metagenome , Adolescent , Bacteria/genetics , Child , Child, Preschool , Female , Fungi/genetics , High-Throughput Nucleotide Sequencing , Humans , Lung Diseases/diagnosis , Male , Metagenomics , Microbiota , Missed Diagnosis , Pilot Projects , Retrospective Studies , Viruses/genetics
15.
Proc Natl Acad Sci U S A ; 115(52): E12353-E12362, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30482864

ABSTRACT

Lower respiratory tract infections (LRTIs) lead to more deaths each year than any other infectious disease category. Despite this, etiologic LRTI pathogens are infrequently identified due to limitations of existing microbiologic tests. In critically ill patients, noninfectious inflammatory syndromes resembling LRTIs further complicate diagnosis. To address the need for improved LRTI diagnostics, we performed metagenomic next-generation sequencing (mNGS) on tracheal aspirates from 92 adults with acute respiratory failure and simultaneously assessed pathogens, the airway microbiome, and the host transcriptome. To differentiate pathogens from respiratory commensals, we developed a rules-based model (RBM) and logistic regression model (LRM) in a derivation cohort of 20 patients with LRTIs or noninfectious acute respiratory illnesses. When tested in an independent validation cohort of 24 patients, both models achieved accuracies of 95.5%. We next developed pathogen, microbiome diversity, and host gene expression metrics to identify LRTI-positive patients and differentiate them from critically ill controls with noninfectious acute respiratory illnesses. When tested in the validation cohort, the pathogen metric performed with an area under the receiver-operating curve (AUC) of 0.96 (95% CI, 0.86-1.00), the diversity metric with an AUC of 0.80 (95% CI, 0.63-0.98), and the host transcriptional classifier with an AUC of 0.88 (95% CI, 0.75-1.00). Combining these achieved a negative predictive value of 100%. This study suggests that a single streamlined protocol offering an integrated genomic portrait of pathogen, microbiome, and host transcriptome may hold promise as a tool for LRTI diagnosis.


Subject(s)
Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/immunology , Sequence Analysis, DNA/methods , Adult , Aged , Aged, 80 and over , Area Under Curve , Case-Control Studies , Cohort Studies , Critical Illness , Female , Humans , Male , Microbiota/genetics , Middle Aged , Predictive Value of Tests , Respiratory Tract Infections/microbiology , Transcriptome/genetics , Whole Genome Sequencing/methods
16.
Mod Pathol ; 30(8): 1086-1099, 2017 08.
Article in English | MEDLINE | ID: mdl-28548128

ABSTRACT

Secretory carcinomas of the breast are rare tumors with distinct histologic features, recurrent t(12;15)(p13;q25) translocation resulting in ETV6-NTRK3 gene fusion and indolent clinical behavior. Mammary analog secretory carcinomas arising in other sites are histopathologically similar to the breast tumors and also harbor ETV6-NTRK3 fusions. Breast secretory carcinomas are often triple (estrogen and progesterone receptor, HER2) negative with a basal-like immunophenotype. However, genomic studies are lacking, and whether these tumors share genetic features with other basal and/or triple negative breast cancers is unknown. Aside from shared ETV6-NTRK3 fusions, the genetic relatedness of secretory carcinomas arising in different sites is also uncertain. We immunoprofiled and sequenced 510 cancer-related genes in nine breast secretory carcinomas and six salivary gland mammary analog secretory carcinomas. Immunoprofiles of breast and salivary gland secretory carcinomas were similar. All the tumors showed strong diffuse MUC4 expression (n=15), and SOX10 was positive in all nine breast and in five out of six salivary gland tumors. All breast secretory carcinomas were triple negative or weakly ER-positive, and all tumors at both the sites expressed CK5/6 and/or EGFR, consistent with a basal-like phenotype. Sequencing revealed classic ETV6-NTRK3 fusion genes in all cases, including in carcinoma in situ of one breast tumor. Translocations were reciprocal and balanced in six out of nine breast and three out of six salivary gland tumors and were complex in three others. In contrast to most breast basal carcinomas, the mutational burden of secretory carcinomas was very low, and no additional pathogenic aberrations were identified in genes typically mutated in breast cancer. Five (56%) breast and two (33%) salivary gland tumors had simple genomes without copy number changes; the remainder had very few changes, averaging 1.3 per tumor. The ETV6-NTRK3 derivative chromosome was duplicated in one breast and one salivary gland tumor, and was the only copy number change in the latter. The findings highlight breast secretory carcinoma as a subtype more closely related to mammary analog secretory carcinoma than to basal/triple negative breast cancers of no special type. Lack of pathogenic mutations in common cancer-related genes suggests that ETV6-NTRK3 alone may suffice to drive these tumors and likely helps explain their indolent behavior.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Mammary Analogue Secretory Carcinoma/genetics , Adolescent , Adult , Aged , Breast Neoplasms/pathology , Female , Gene Expression Profiling , Humans , Male , Mammary Analogue Secretory Carcinoma/pathology , Middle Aged , Oncogene Proteins, Fusion/genetics , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/pathology , Young Adult
17.
PLoS One ; 12(5): e0178163, 2017.
Article in English | MEDLINE | ID: mdl-28542423

ABSTRACT

Genetic manipulation of the deadly malaria parasite Plasmodium falciparum remains challenging, but the rise of CRISPR/Cas9-based genome editing tools is increasing the feasibility of altering this parasite's genome in order to study its biology. Of particular interest is the investigation of drug targets and drug resistance mechanisms, which have major implications for fighting malaria. We present a new method for introducing drug resistance mutations in P. falciparum without the use of plasmids or the need for cloning homologous recombination templates. We demonstrate this method by introducing edits into the sodium efflux channel PfATP4 by transfection of a purified CRISPR/Cas9-guide RNA ribonucleoprotein complex and a 200-nucleotide single-stranded oligodeoxynucleotide (ssODN) repair template. Analysis of whole genome sequencing data with the variant-finding program MinorityReport confirmed that only the intended edits were made, and growth inhibition assays confirmed that these mutations confer resistance to the antimalarial SJ733. The method described here is ideally suited for the introduction of mutations that confer a fitness advantage under selection conditions, and the novel finding that an ssODN can function as a repair template in P. falciparum could greatly simplify future editing attempts regardless of the nuclease used or the delivery method.


Subject(s)
CRISPR-Cas Systems/genetics , Drug Resistance/genetics , Genome, Protozoan , H(+)-K(+)-Exchanging ATPase/genetics , Isoquinolines/pharmacology , Mutation/genetics , Plasmodium falciparum/genetics , Amino Acid Sequence , Base Sequence , Genetic Vectors , Humans , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Plasmids , RNA Editing/genetics
18.
Cell Rep ; 18(2): 454-467, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28076789

ABSTRACT

Low-complexity "prion-like" domains in key RNA-binding proteins (RBPs) mediate the reversible assembly of RNA granules. Individual RBPs harboring these domains have been linked to specific neurodegenerative diseases. Although their aggregation in neurodegeneration has been extensively characterized, it remains unknown how the process of aging disturbs RBP dynamics. We show that a wide variety of RNA granule components, including stress granule proteins, become highly insoluble with age in C. elegans and that reduced insulin/insulin-like growth factor 1 (IGF-1) daf-2 receptor signaling efficiently prevents their aggregation. Importantly, stress-granule-related RBP aggregates are associated with reduced fitness. We show that heat shock transcription factor 1 (HSF-1) is a main regulator of stress-granule-related RBP aggregation in both young and aged animals. During aging, increasing DAF-16 activity restores dynamic stress-granule-related RBPs, partly by decreasing the buildup of other misfolded proteins that seed RBP aggregation. Longevity-associated mechanisms found to maintain dynamic RBPs during aging could be relevant for neurodegenerative diseases.


Subject(s)
Aging/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cytoplasmic Granules/metabolism , Heat-Shock Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Signal Transduction , Animals , Longevity , Mutation/genetics , Protein Aggregates , RNA/metabolism , RNA-Binding Proteins/metabolism , Receptor, Insulin/metabolism , Solubility
20.
Genome Med ; 8(1): 90, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27562436

ABSTRACT

BACKGROUND: Ocular infections remain a major cause of blindness and morbidity worldwide. While prognosis is dependent on the timing and accuracy of diagnosis, the etiology remains elusive in ~50 % of presumed infectious uveitis cases. The objective of this study is to determine if unbiased metagenomic deep sequencing (MDS) can accurately detect pathogens in intraocular fluid samples of patients with uveitis. METHODS: This is a proof-of-concept study, in which intraocular fluid samples were obtained from five subjects with known diagnoses, and one subject with bilateral chronic uveitis without a known etiology. Samples were subjected to MDS, and results were compared with those from conventional diagnostic tests. Pathogens were identified using a rapid computational pipeline to analyze the non-host sequences obtained from MDS. RESULTS: Unbiased MDS of intraocular fluid produced results concordant with known diagnoses in subjects with (n = 4) and without (n = 1) uveitis. Samples positive for Cryptococcus neoformans, Toxoplasma gondii, and herpes simplex virus 1 as tested by a Clinical Laboratory Improvement Amendments-certified laboratory were correctly identified with MDS. Rubella virus was identified in one case of chronic bilateral idiopathic uveitis. The subject's strain was most closely related to a German rubella virus strain isolated in 1992, one year before he developed a fever and rash while living in Germany. The pattern and the number of viral identified mutations present in the patient's strain were consistent with long-term viral replication in the eye. CONCLUSIONS: MDS can identify fungi, parasites, and DNA and RNA viruses in minute volumes of intraocular fluid samples. The identification of chronic intraocular rubella virus infection highlights the eye's role as a long-term pathogen reservoir, which has implications for virus eradication and emerging global epidemics.


Subject(s)
Cryptococcus neoformans/genetics , Herpesvirus 1, Human/genetics , Metagenomics , Rubella virus/genetics , Toxoplasma/genetics , Uveitis/diagnosis , Aqueous Humor/microbiology , Aqueous Humor/parasitology , Aqueous Humor/virology , Cryptococcosis/diagnosis , Cryptococcosis/microbiology , Cryptococcus neoformans/pathogenicity , Herpes Simplex/diagnosis , Herpes Simplex/virology , Herpesvirus 1, Human/pathogenicity , High-Throughput Nucleotide Sequencing , Humans , Phylogeny , Rubella/diagnosis , Rubella/virology , Rubella virus/classification , Rubella virus/pathogenicity , Toxoplasma/pathogenicity , Toxoplasmosis/diagnosis , Toxoplasmosis/parasitology , Uvea/microbiology , Uvea/parasitology , Uvea/pathology , Uvea/virology , Uveitis/microbiology , Uveitis/parasitology , Uveitis/virology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL