1
|
Aalam J, Ahmad Shah SN, Parveen R. An extensive review on infectious disease diagnosis using machine learning techniques and next generation sequencing: State-of-the-art and perspectives. Comput Biol Med 2025; 189:109962. [PMID: 40054170 DOI: 10.1016/j.compbiomed.2025.109962] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 04/01/2025]
Abstract
Infectious diseases, including tuberculosis (TB), HIV/AIDS, and emerging pathogens like COVID-19 pose severe global health challenges due to their rapid spread and significant morbidity and mortality rates. Next-generation sequencing (NGS) and machine learning (ML) have emerged as transformative technologies for enhancing disease diagnosis and management. OBJECTIVE This review aims to explore integrating ML techniques with NGS for diagnosing infectious diseases, highlighting their effectiveness and identifying existing challenges. METHODS A comprehensive literature review spanning the past decade was conducted using reputable databases, including IEEE Xplore, PubMed, Scopus, SpringerLink, and Science Direct. Research papers, articles, and conference proceedings meeting stringent quality criteria were analysed to assess the performance of ML algorithms applied to NGS and metagenomic NGS (mNGS) data. RESULTS The findings reveal that ML algorithms, such as deep neural networks (DNNs), support vector machines (SVM), and K-nearest neighbours (KNN), achieve high accuracy rates, often exceeding 95 %, in diagnosing infectious diseases. Deep learning methods excel in genomic and metagenomic data analysis, while traditional algorithms like Gaussian mixture models (GMM) also demonstrate robust classification capabilities. Challenges include reliance on single data types and difficulty distinguishing closely related pathogens. CONCLUSION The integration of ML and NGS significantly advances infectious disease diagnosis, offering rapid and precise detection capabilities. Addressing current limitations can further enhance the effectiveness of these technologies, ultimately improving global public health outcomes.
Collapse
Affiliation(s)
- Javed Aalam
- Department of Computer Science, Jamia Millia Islamia, New Delhi, 110025, India.
| | | | - Rafat Parveen
- Department of Computer Science, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
2
|
Verstockt S, Hannes L, Jans DS, Deman S, Souche E, van der Werf I, Vandermeulen L, Lobaton T, Laukens D, Verstockt B, Van Houdt J, Hoischen A, Vermeire S, Cleynen I. MIP4IBD: An Easy and Rapid Genotyping-by-Sequencing Assay for the Inflammatory Bowel Diseases Risk Loci. Inflamm Bowel Dis 2025; 31:786-799. [PMID: 39657915 DOI: 10.1093/ibd/izae289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Inflammatory bowel diseases (IBD) are polygenic, with many genetic variants contributing to disease risk. Knowing the genotype of specific variants or calculating a combined genetic risk score benefits translational and functional research. To address this, we developed MIP4IBD, a flexible and cost-effective genotyping-by-sequencing assay using molecular inversion probes (MIPs). METHODS The assay targets 463 IBD risk variants, and 77 additional relevant variants. Molecular inversion probes capture and library preparation were optimized using 15 IBD DNA samples, comparing genotypes with immunochip. A custom GitHub pipeline was created for data processing, performance testing, and genotype calling. The final design was validated on a larger scale (149 IBD patients, 104 non-IBD controls, and 3 external cell lines), incorporating post hoc quality control criteria. RESULTS The assay achieved a 3.5-day turnaround time at €15 per sample with optimal sample throughput, demonstrating a 92.6% success rate in variant capture and genotype concordance rates of 99.3% and 99.6% with Infinium Global Screening Array24 BeadChip and WGS, respectively. A downstream application involved the calculation of a weighted IBD polygenic risk score (PRS), which was significantly higher in IBD patients than controls (mean 0.42 vs -0.49, P = 1.95E-11). Individuals in the highest PRS quartile had a 15.7-fold (95% CI: 6.5-38.3) risk of developing IBD and an earlier age of onset (26 vs 37 years, P = 0.02), compared to the lowest quartile. CONCLUSIONS MIP4IBD is a validated, scalable genotyping assay targeting IBD risk loci, with an integrated bioinformatics pipeline from sequencing data to genotypes and PRS calculation. Its cost-effectiveness and flexibility for additional variants make it particularly appealing for translational and clinical applications.
Collapse
Affiliation(s)
- Sare Verstockt
- Department of Chronic Diseases and Metabolism (CHROMETA), University of Leuven, Herestraat 49 Box 701, 3000 Leuven, Belgium
| | - Laurens Hannes
- Department of Human Genetics, University of Leuven, KU Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
- Center for Human Genetics, University Hospitals Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
| | - Deborah Sarah Jans
- Department of Human Genetics, Laboratory for Complex Genetics Leuven, University of Leuven, Herestraat 49 Box 604, 3000 Leuven, Belgium
| | - Stephanie Deman
- Department of Human Genetics, University of Leuven, KU Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
- Genomics Core, University Hospitals Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
| | - Erika Souche
- Department of Human Genetics, University of Leuven, KU Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
- Genomics Core, University Hospitals Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
| | - Ilse van der Werf
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Liv Vandermeulen
- Department of Gastroenterology, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Triana Lobaton
- Department of Gastroenterology, University Hospital of Ghent, Corneel Heymanslaan 10, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Debby Laukens
- Department of Internal Medicine and Pediatrics, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Bram Verstockt
- Department of Chronic Diseases and Metabolism (CHROMETA), University of Leuven, Herestraat 49 Box 701, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Jeroen Van Houdt
- J&J Innovative Medicine, Antwerpseweg 15-17, 2340 Beerse, Belgium
| | - Alexander Hoischen
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Department of Human Genetics, Heyendaalseweg 135 Box 9010, 6525 AJ Nijmegen, The Netherlands
| | - Séverine Vermeire
- Department of Chronic Diseases and Metabolism (CHROMETA), University of Leuven, Herestraat 49 Box 701, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Isabelle Cleynen
- Department of Human Genetics, Laboratory for Complex Genetics Leuven, University of Leuven, Herestraat 49 Box 604, 3000 Leuven, Belgium
| |
Collapse
|
3
|
Sun Z, Liu J, Zhang M, Wang T, Huang S, Weiss ST, Liu YY. Removal of false positives in metagenomics-based taxonomy profiling via targeting Type IIB restriction sites. Nat Commun 2023; 14:5321. [PMID: 37658057 PMCID: PMC10474111 DOI: 10.1038/s41467-023-41099-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/22/2023] [Indexed: 09/03/2023] Open
Abstract
Accurate species identification and abundance estimation are critical for the interpretation of whole metagenome sequencing (WMS) data. Yet, existing metagenomic profilers suffer from false-positive identifications, which can account for more than 90% of total identified species. Here, by leveraging species-specific Type IIB restriction endonuclease digestion sites as reference instead of universal markers or whole microbial genomes, we present a metagenomic profiler, MAP2B (MetAgenomic Profiler based on type IIB restriction sites), to resolve those issues. We first illustrate the pitfalls of using relative abundance as the only feature in determining false positives. We then propose a feature set to distinguish false positives from true positives, and using simulated metagenomes from CAMI2, we establish a false-positive recognition model. By benchmarking the performance in metagenomic profiling using a simulation dataset with varying sequencing depth and species richness, we illustrate the superior performance of MAP2B over existing metagenomic profilers in species identification. We further test the performance of MAP2B using real WMS data from an ATCC mock community, confirming its superior precision against sequencing depth. Finally, by leveraging WMS data from an IBD cohort, we demonstrate the taxonomic features generated by MAP2B can better discriminate IBD and predict metabolomic profiles.
Collapse
Affiliation(s)
- Zheng Sun
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jiang Liu
- Qingdao OE Biotechnology Company Limited, Qingdao, Shandong, China
| | - Meng Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Tong Wang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shi Huang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Center for Artificial Intelligence and Modeling, The Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
| |
Collapse
|
4
|
Hernández-Neuta I, Magoulopoulou A, Pineiro F, Lisby JG, Gulberg M, Nilsson M. Highly multiplexed targeted sequencing strategy for infectious disease surveillance. BMC Biotechnol 2023; 23:31. [PMID: 37612665 PMCID: PMC10463907 DOI: 10.1186/s12896-023-00804-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Global efforts to characterize diseases of poverty are hampered by lack of affordable and comprehensive detection platforms, resulting in suboptimal allocation of health care resources and inefficient disease control. Next generation sequencing (NGS) can provide accurate data and high throughput. However, shotgun and metagenome-based NGS approaches are limited by low concentrations of microbial DNA in clinical samples, requirements for tailored sample and library preparations plus extensive bioinformatics analysis. Here, we adapted molecular inversion probes (MIPs) as a cost-effective target enrichment approach to characterize microbial infections from blood samples using short-read sequencing. We designed a probe panel targeting 2 bacterial genera, 21 bacterial and 6 fungi species and 7 antimicrobial resistance markers (AMRs). RESULTS Our approach proved to be highly specific to detect down to 1 in a 1000 pathogen DNA targets contained in host DNA. Additionally, we were able to accurately survey pathogens and AMRs in 20 out of 24 samples previously profiled with routine blood culture for sepsis. CONCLUSIONS Overall, our targeted assay identifies microbial pathogens and AMRs with high specificity at high throughput, without the need for extensive sample preparation or bioinformatics analysis, simplifying its application for characterization and surveillance of infectious diseases in medium- to low- resource settings.
Collapse
Affiliation(s)
- Iván Hernández-Neuta
- Department of Biochemistry and Biophysics, Faculty of Science, Stockholm University, Svante Arrhenius väg 16C, Stockholm, 104 05, Sweden
- Science for Life Laboratory (SciLifeLab), Tomtebodavägen 23, 171 65, Solna, Sweden
| | - Anastasia Magoulopoulou
- Department of Biochemistry and Biophysics, Faculty of Science, Stockholm University, Svante Arrhenius väg 16C, Stockholm, 104 05, Sweden
- Science for Life Laboratory (SciLifeLab), Tomtebodavägen 23, 171 65, Solna, Sweden
| | - Flor Pineiro
- Department of Biochemistry and Biophysics, Faculty of Science, Stockholm University, Svante Arrhenius väg 16C, Stockholm, 104 05, Sweden
- Science for Life Laboratory (SciLifeLab), Tomtebodavägen 23, 171 65, Solna, Sweden
| | - Jan Gorm Lisby
- Department of Clinical Microbiology, Amager and Hvidovre Hospital, University of Copenhagen, Kettegaard Alle 30, Hvidovre, 2650, Denmark
| | - Mats Gulberg
- Q-linea AB, Dag Hammarskjölds Väg 52A, Uppsala, 752 37, Sweden
| | - Mats Nilsson
- Department of Biochemistry and Biophysics, Faculty of Science, Stockholm University, Svante Arrhenius väg 16C, Stockholm, 104 05, Sweden.
- Science for Life Laboratory (SciLifeLab), Tomtebodavägen 23, 171 65, Solna, Sweden.
| |
Collapse
|
5
|
Chen Y, Mao L, Lai D, Xu W, Zhang Y, Wu S, Yang D, Zhao S, Liu Z, Xiao Y, Tang Y, Meng X, Wang M, Shi J, Chen Q, Shu Q. Improved targeting of the 16S rDNA nanopore sequencing method enables rapid pathogen identification in bacterial pneumonia in children. Front Cell Infect Microbiol 2023; 12:1001607. [PMID: 36699719 PMCID: PMC9868273 DOI: 10.3389/fcimb.2022.1001607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Objectives To develop a rapid and low-cost method for 16S rDNA nanopore sequencing. Methods This was a prospective study on a 16S rDNA nanopore sequencing method. We developed this nanopore barcoding 16S sequencing method by adding barcodes to the 16S primer to reduce the reagent cost and simplify the experimental procedure. Twenty-one common pulmonary bacteria (7 reference strains, 14 clinical isolates) and 94 samples of bronchoalveolar lavage fluid from children with severe pneumonia were tested. Results indicating low-abundance pathogenic bacteria were verified with the polymerase chain reaction (PCR). Further, the results were compared with those of culture or PCR. Results The turnaround time was shortened to 6~8 hours and the reagent cost of DNA preparation was reduced by employing a single reaction adding barcodes to the 16S primer in advance. The accuracy rate for the 21 common pulmonary pathogens with an abundance ≥ 99% was 100%. Applying the culture or PCR results as the gold standard, 71 (75.5%) of the 94 patients were positive, including 25 positive cultures (26.6%) and 52 positive quantitative PCRs (55.3%). The median abundance in the positive culture and qPCR samples were 29.9% and 6.7%, respectively. With an abundance threshold increase of 1%, 5%, 10%, 15% and 20%, the test sensitivity decreased gradually to 98.6%, 84.9%, 72.6%, 67.1% and 64.4%, respectively, and the test specificity increased gradually to 33.3%, 71.4%, 81.0%, 90.5% and 100.0%, respectively. Conclusions The nanopore barcoding 16S sequencing method can rapidly identify the pathogens causing bacterial pneumonia in children.
Collapse
Affiliation(s)
- Yinghu Chen
- The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Clinical Research Center for Child Health, Hangzhou, China,Joint Research Center for Molecular Diagnosis of Severe Infection in Children, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Lingfeng Mao
- Joint Research Center for Molecular Diagnosis of Severe Infection in Children, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Dengming Lai
- The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Clinical Research Center for Child Health, Hangzhou, China,Joint Research Center for Molecular Diagnosis of Severe Infection in Children, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Weize Xu
- The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Clinical Research Center for Child Health, Hangzhou, China,Joint Research Center for Molecular Diagnosis of Severe Infection in Children, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Yuebai Zhang
- The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Clinical Research Center for Child Health, Hangzhou, China
| | - Sihao Wu
- Joint Research Center for Molecular Diagnosis of Severe Infection in Children, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Di Yang
- Joint Research Center for Molecular Diagnosis of Severe Infection in Children, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Shaobo Zhao
- The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Clinical Research Center for Child Health, Hangzhou, China
| | - Zhicong Liu
- The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Clinical Research Center for Child Health, Hangzhou, China
| | - Yi Xiao
- The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Clinical Research Center for Child Health, Hangzhou, China
| | - Yi Tang
- Joint Research Center for Molecular Diagnosis of Severe Infection in Children, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Xiaofang Meng
- Joint Research Center for Molecular Diagnosis of Severe Infection in Children, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Min Wang
- Joint Research Center for Molecular Diagnosis of Severe Infection in Children, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Jueliang Shi
- Joint Research Center for Molecular Diagnosis of Severe Infection in Children, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Qixing Chen
- The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Clinical Research Center for Child Health, Hangzhou, China,Joint Research Center for Molecular Diagnosis of Severe Infection in Children, Binjiang Institute of Zhejiang University, Hangzhou, China,*Correspondence: Qixing Chen, ; Qiang Shu,
| | - Qiang Shu
- The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Clinical Research Center for Child Health, Hangzhou, China,Joint Research Center for Molecular Diagnosis of Severe Infection in Children, Binjiang Institute of Zhejiang University, Hangzhou, China,*Correspondence: Qixing Chen, ; Qiang Shu,
| |
Collapse
|
6
|
Stefan CP, Hall AT, Graham AS, Minogue TD. Comparison of Illumina and Oxford Nanopore Sequencing Technologies for Pathogen Detection from Clinical Matrices Using Molecular Inversion Probes. J Mol Diagn 2022; 24:395-405. [PMID: 35085783 DOI: 10.1016/j.jmoldx.2021.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/19/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Next-generation sequencing is rapidly finding footholds in numerous microbiological fields, including infectious disease diagnostics. Here, we describe a molecular inversion probe panel for the identification of bacterial, viral, and parasitic pathogens. We describe the ability of Illumina and Oxford Nanopore Technologies (ONT) to sequence small amplicons originating from this panel for the identification of pathogens in complex matrices. The panel correctly classified 31 bacterial pathogens directly from positive blood culture bottles with a genus-level concordance of 96.7% and 90.3% on the Illumina and ONT platforms, respectively. Both sequencing platforms detected 18 viral and parasitic organisms directly from mock clinical samples of plasma and whole blood at concentrations of 104 PFU/mL with few exceptions. In general, Illumina sequencing exhibited greater read counts with lower percent mapped reads; however, this resulted in no effect on limits of detection compared with ONT sequencing. Mock clinical evaluation of the probe panel on the Illumina and ONT platforms resulted in positive predictive values of 0.91 and 0.88 and negative predictive values of 1 and 1 from de-identified human chikungunya virus samples compared with gold standard quantitative RT-PCR. Overall, these data show that molecular inversion probes are an adaptable technology capable of pathogen detection from complex sample matrices on current next-generation sequencing platforms.
Collapse
Affiliation(s)
- Christopher P Stefan
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland
| | - Adrienne T Hall
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland
| | - Amanda S Graham
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland
| | - Timothy D Minogue
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland.
| |
Collapse
|
7
|
RESCRIPt: Reproducible sequence taxonomy reference database management. PLoS Comput Biol 2021; 17:e1009581. [PMID: 34748542 PMCID: PMC8601625 DOI: 10.1371/journal.pcbi.1009581] [Citation(s) in RCA: 340] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 11/18/2021] [Accepted: 10/21/2021] [Indexed: 12/22/2022] Open
Abstract
Nucleotide sequence and taxonomy reference databases are critical resources for widespread applications including marker-gene and metagenome sequencing for microbiome analysis, diet metabarcoding, and environmental DNA (eDNA) surveys. Reproducibly generating, managing, using, and evaluating nucleotide sequence and taxonomy reference databases creates a significant bottleneck for researchers aiming to generate custom sequence databases. Furthermore, database composition drastically influences results, and lack of standardization limits cross-study comparisons. To address these challenges, we developed RESCRIPt, a Python 3 software package and QIIME 2 plugin for reproducible generation and management of reference sequence taxonomy databases, including dedicated functions that streamline creating databases from popular sources, and functions for evaluating, comparing, and interactively exploring qualitative and quantitative characteristics across reference databases. To highlight the breadth and capabilities of RESCRIPt, we provide several examples for working with popular databases for microbiome profiling (SILVA, Greengenes, NCBI-RefSeq, GTDB), eDNA and diet metabarcoding surveys (BOLD, GenBank), as well as for genome comparison. We show that bigger is not always better, and reference databases with standardized taxonomies and those that focus on type strains have quantitative advantages, though may not be appropriate for all use cases. Most databases appear to benefit from some curation (quality filtering), though sequence clustering appears detrimental to database quality. Finally, we demonstrate the breadth and extensibility of RESCRIPt for reproducible workflows with a comparison of global hepatitis genomes. RESCRIPt provides tools to democratize the process of reference database acquisition and management, enabling researchers to reproducibly and transparently create reference materials for diverse research applications. RESCRIPt is released under a permissive BSD-3 license at https://github.com/bokulich-lab/RESCRIPt. Generating and managing sequence and taxonomy reference data presents a bottleneck to many researchers, whether they are generating custom databases or attempting to format existing, curated reference databases for use with standard sequence analysis tools. Evaluating database quality and choosing the “best” database can be an equally formidable challenge. We developed RESCRIPt to alleviate this bottleneck, supporting reproducible, streamlined generation, curation, and evaluation of reference sequence databases. RESCRIPt uses QIIME 2 artifact file formats, which store all processing steps as data provenance within each file, allowing researchers to retrace the computational steps used to generate any given file. We used RESCRIPt to benchmark several commonly used marker-gene sequence databases for 16S rRNA genes, ITS, and COI sequences, demonstrating both the utility of RESCRIPt to streamline use of these databases, but also to evaluate several qualitative and quantitative characteristics of each database. We show that larger databases are not always best, and curation steps to reduce redundancy and filter out noisy sequences may be beneficial for some applications. We anticipate that RESCRIPt will streamline the use, management, and evaluation/selection of reference database materials for microbiomics, diet metabarcoding, eDNA, and other diverse applications.
Collapse
|
8
|
Enhancement of Antibiofilm Activity of Ciprofloxacin against Staphylococcus aureus by Administration of Antimicrobial Peptides. Antibiotics (Basel) 2021; 10:antibiotics10101159. [PMID: 34680739 PMCID: PMC8532819 DOI: 10.3390/antibiotics10101159] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 11/17/2022] Open
Abstract
Staphylococcus aureus can develop resistance by mutation, transfection or biofilm formation. Resistance was induced in S. aureus by growth in sub-inhibitory concentrations of ciprofloxacin for 30 days. The ability of the antimicrobials to disrupt biofilms was determined using crystal violet and live/dead staining. Effects on the cell membranes of biofilm cells were evaluated by measuring release of dyes and ATP, and nucleic acids. None of the strains developed resistance to AMPs while only S. aureus ATCC 25923 developed resistance (128 times) to ciprofloxacin after 30 passages. Only peptides reduced biofilms of ciprofloxacin-resistant cells. The antibiofilm effect of melimine with ciprofloxacin was more (27%) than with melimine alone at 1X MIC (p < 0.001). Similarly, at 1X MIC the combination of Mel4 and ciprofloxacin produced more (48%) biofilm disruption than Mel4 alone (p < 0.001). Combinations of either of the peptides with ciprofloxacin at 2X MIC released ≥ 66 nM ATP, more than either peptide alone (p ≤ 0.005). At 2X MIC, only melimine in combination with ciprofloxacin released DNA/RNA which was three times more than that released by melimine alone (p = 0.043). These results suggest the potential use of melimine and Mel4 with conventional antibiotics for the treatment of S. aureus biofilms.
Collapse
|
9
|
Qian L, Shi Y, Li F, Wang Y, Ma M, Zhang Y, Shao YW, Zheng G, Zhang G. Metagenomic Next-Generation Sequencing of Cerebrospinal Fluid for the Diagnosis of External Ventricular and Lumbar Drainage-Associated Ventriculitis and Meningitis. Front Microbiol 2021; 11:596175. [PMID: 33381092 PMCID: PMC7767851 DOI: 10.3389/fmicb.2020.596175] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/13/2020] [Indexed: 12/20/2022] Open
Abstract
Metagenomic next-generation sequencing (mNGS) has become a widely used technology that can accurately detect individual pathogens. This prospective study was performed between February 2019 and September 2019 in one of the largest clinical neurosurgery centers in China. The study aimed to evaluate the performance of mNGS on cerebrospinal fluid (CSF) from neurosurgical patients for the diagnosis of external ventricular and lumbar drainage (EVD/LD)-associated ventriculitis and meningitis (VM). We collected CSF specimens from neurosurgical patients with EVD/LD for more than 24 h to perform conventional microbiological studies and mNGS analyses in a pairwise manner. We also investigated the usefulness of mNGS of CSF for the diagnosis of EVD/LD-associated VM. In total, 102 patients were enrolled in this study and divided into three groups, including confirmed VM (cVM) (39), suspected VM (sVM) (49), and non-VM (nVM) (14) groups. Of all the patients, mNGS detected 21 Gram-positive bacteria, 20 Gram-negative bacteria, and five fungi. The three primary bacteria detected were Staphylococcus epidermidis (9), Acinetobacter baumannii (5), and Staphylococcus aureus (3). The mNGS-positive coincidence rate of confirmed EVD/LD-associated VM was 61.54% (24/39), and the negative coincidence rate of the nVM group was 100% (14/14). Of 15 VM pathogens not identified by mNGS in the cVM group, eight were negative with mNGS and seven were inconsistent with the conventional microbiological identification results. In addition, mNGS identified pathogens in 22 cases that were negative using conventional methods; of them, 10 patients received a favorable clinical treatment; thus, showing the benefit of mNGS-guided therapy.
Collapse
Affiliation(s)
- Lingye Qian
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yijun Shi
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fangqiang Li
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yufei Wang
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Miao Ma
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yanfang Zhang
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yang W Shao
- Nanjing Geneseeq Technology Inc., Nanjing, China.,School of Public Health, Nanjing Medical University, Nanjing, China
| | - Guanghui Zheng
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guojun Zhang
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Anis E, Ilha MRS, Engiles JB, Wilkes RP. Evaluation of targeted next-generation sequencing for detection of equine pathogens in clinical samples. J Vet Diagn Invest 2020; 33:227-234. [PMID: 33305693 DOI: 10.1177/1040638720978381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Equine infectious disease outbreaks may have profound economic impact, resulting in losses of millions of dollars of revenue as a result of horse loss, quarantine, and cancelled events. Early and accurate diagnosis is essential to limit the spread of infectious diseases. However, laboratory detection of infectious agents, especially the simultaneous detection of multiple agents, can be challenging to the clinician and diagnostic laboratory. Next-generation sequencing (NGS), which allows millions of DNA templates to be sequenced simultaneously in a single reaction, is an ideal technology for comprehensive testing. We conducted a proof-of-concept study of targeted NGS to detect 62 common equine bacterial, viral, and parasitic pathogens in clinical samples. We designed 264 primers and constructed a bioinformatics tool for the detection of targeted pathogens. The designed primers were able to specifically detect the intended pathogens. Results of testing 27 clinical samples with our targeted NGS assay compared with results of routine tests (assessed as a group) yielded positive percent agreement of 81% and negative percent agreement of 83%, overall agreement of 81%, and kappa of 0.56 (moderate agreement). This moderate agreement was likely the result of low sensitivity of some primers. However, our NGS assay successfully detected multiple pathogens in the clinical samples, including some pathogens missed by routine techniques.
Collapse
Affiliation(s)
- Eman Anis
- Department of Pathobiology, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA.,Department of Virology, Faculty of Veterinary Medicine, University of Sadat, El Beheira Governorate, Sadat City, Egypt
| | - Marcia R S Ilha
- Tifton Veterinary Diagnostic and Investigational Laboratory, College of Veterinary Medicine, University of Georgia, Tifton, GA
| | - Julie B Engiles
- Department of Pathobiology, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA
| | - Rebecca P Wilkes
- Tifton Veterinary Diagnostic and Investigational Laboratory, College of Veterinary Medicine, University of Georgia, Tifton, GA.,Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN
| |
Collapse
|
11
|
Kim J, Ahn JK, Kim JS, Choi BR, Cho J, Lee H. Highly selective detection of single nucleotide polymorphism (SNP) using a dumbbell DNA probe with a gap-filling approach. J IND ENG CHEM 2020. [DOI: 10.1016/j.jiec.2020.03.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
12
|
A Survey of Antimicrobial Resistance Determinants in Category A Select Agents, Exempt Strains, and Near-Neighbor Species. Int J Mol Sci 2020; 21:ijms21051669. [PMID: 32121349 PMCID: PMC7084191 DOI: 10.3390/ijms21051669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 01/02/2023] Open
Abstract
A dramatic increase in global antimicrobial resistance (AMR) has been well documented. Of particular concern is the dearth of information regarding the spectrum and prevalence of AMR within Category A Select Agents. Here, we performed a survey of horizontally and vertically transferred AMR determinants among Category A agents and their near neighbors. Microarrays provided broad spectrum screening of 127 Francisella spp., Yersinia spp., and Bacillus spp. strains for the presence/absence of 500+ AMR genes (or families of genes). Detecting a broad variety of AMR genes in each genus, microarray analysis also picked up the presence of an engineered plasmid in a Y. pestis strain. High resolution melt analysis (HRMA) was also used to assess the presence of quinolone resistance-associated mutations in 100 of these strains. Though HRMA was able to detect resistance-causing point mutations in B. anthracis strains, it was not capable of discriminating these point mutations from other nucleotide substitutions (e.g., arising from sequence differences in near neighbors). Though these technologies are well-established, to our knowledge, this is the largest survey of Category A agents and their near-neighbor species for genes covering multiple mechanisms of AMR.
Collapse
|
13
|
Sichtig H, Minogue T, Yan Y, Stefan C, Hall A, Tallon L, Sadzewicz L, Nadendla S, Klimke W, Hatcher E, Shumway M, Aldea DL, Allen J, Koehler J, Slezak T, Lovell S, Schoepp R, Scherf U. FDA-ARGOS is a database with public quality-controlled reference genomes for diagnostic use and regulatory science. Nat Commun 2019; 10:3313. [PMID: 31346170 PMCID: PMC6658474 DOI: 10.1038/s41467-019-11306-6] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 07/02/2019] [Indexed: 02/08/2023] Open
Abstract
FDA proactively invests in tools to support innovation of emerging technologies, such as infectious disease next generation sequencing (ID-NGS). Here, we introduce FDA-ARGOS quality-controlled reference genomes as a public database for diagnostic purposes and demonstrate its utility on the example of two use cases. We provide quality control metrics for the FDA-ARGOS genomic database resource and outline the need for genome quality gap filling in the public domain. In the first use case, we show more accurate microbial identification of Enterococcus avium from metagenomic samples with FDA-ARGOS reference genomes compared to non-curated GenBank genomes. In the second use case, we demonstrate the utility of FDA-ARGOS reference genomes for Ebola virus target sequence comparison as part of a composite validation strategy for ID-NGS diagnostic tests. The use of FDA-ARGOS as an in silico target sequence comparator tool combined with representative clinical testing could reduce the burden for completing ID-NGS clinical trials. To be able to use infectious disease next generation sequencing as a diagnostic tool, appropriate reference datasets are required. Here, Sichtig et al. describe FDA-ARGOS, a reference database for high-quality microbial reference genomes, and demonstrate its utility on the example of two use cases.
Collapse
Affiliation(s)
- Heike Sichtig
- U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA.
| | - Timothy Minogue
- U.S. Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, USA.
| | - Yi Yan
- U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Christopher Stefan
- U.S. Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, USA
| | - Adrienne Hall
- U.S. Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, USA
| | - Luke Tallon
- Institute for Genome Sciences at the University of Maryland, 670 W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Lisa Sadzewicz
- Institute for Genome Sciences at the University of Maryland, 670 W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Suvarna Nadendla
- Institute for Genome Sciences at the University of Maryland, 670 W. Baltimore Street, Baltimore, MD, 21201, USA
| | - William Klimke
- National Center for Biotechnology Information, National Library of Medicine, 8600 Rockville Pike, Bethesda, MD, 20894, USA
| | - Eneida Hatcher
- National Center for Biotechnology Information, National Library of Medicine, 8600 Rockville Pike, Bethesda, MD, 20894, USA
| | - Martin Shumway
- National Center for Biotechnology Information, National Library of Medicine, 8600 Rockville Pike, Bethesda, MD, 20894, USA
| | | | - Jonathan Allen
- Lawrence Livermore National Laboratory, P.O. Box 808, Livermore, CA, 94551, USA
| | - Jeffrey Koehler
- U.S. Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, USA
| | - Tom Slezak
- Lawrence Livermore National Laboratory, P.O. Box 808, Livermore, CA, 94551, USA
| | - Stephen Lovell
- U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Randal Schoepp
- U.S. Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, USA
| | - Uwe Scherf
- U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| |
Collapse
|
14
|
Gallon P, Parekh M, Ferrari S, Fasolo A, Ponzin D, Borroni D. Metagenomics in ophthalmology: Hypothesis or real prospective? ACTA ACUST UNITED AC 2019; 23:e00355. [PMID: 31312608 PMCID: PMC6609782 DOI: 10.1016/j.btre.2019.e00355] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/30/2019] [Accepted: 06/22/2019] [Indexed: 12/12/2022]
Abstract
Metagenomic analysis was originally associated with the studies of genetic material from environmental samples. But, with the advent of the Human Microbiome Project, it has now been applied in clinical practices. The ocular surface (OS) is the most exposed part of the eye, colonized by several microbial communities (both, OS and environmental) that contribute to the maintenance of the physiological state. Limited knowledge has been acquired on these microbes due to the limitations of conventional diagnostic methods. Emerging fields of research are focusing on Next Generation Sequencing (NGS) technologies to obtain reliable information on the OS microbiome. Currently only pre-specified pathogens can be detected by conventional culture-based techniques or Polymerase Chain Reaction (PCR), but there are conditions to state whether metagenomics could revolutionize the diagnosis of ocular diseases. The aim of this review is to provide an updated overview of the studies involving NGS technology for OS microbiome.
Collapse
Affiliation(s)
- Paola Gallon
- Fondazione Banca degli Occhi del Veneto, Venice, Italy
| | - Mohit Parekh
- Institute of Ophthalmology, University College London, London, UK
| | | | | | - Diego Ponzin
- Fondazione Banca degli Occhi del Veneto, Venice, Italy
| | - Davide Borroni
- Department of Doctoral Studies, Riga Stradins University, Riga, Latvia
| |
Collapse
|
15
|
Abstract
Clinical metagenomic next-generation sequencing (mNGS), the comprehensive analysis of microbial and host genetic material (DNA and RNA) in samples from patients, is rapidly moving from research to clinical laboratories. This emerging approach is changing how physicians diagnose and treat infectious disease, with applications spanning a wide range of areas, including antimicrobial resistance, the microbiome, human host gene expression (transcriptomics) and oncology. Here, we focus on the challenges of implementing mNGS in the clinical laboratory and address potential solutions for maximizing its impact on patient care and public health.
Collapse
Affiliation(s)
- Charles Y Chiu
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA.
- Department of Medicine, Division of Infectious Diseases, University of California, San Francisco, CA, USA.
| | - Steven A Miller
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
16
|
Liu H, Li PW, Yang WQ, Mi H, Pan JL, Huang YC, Hou ZK, Hou QK, Luo Q, Liu FB. Identification of non-invasive biomarkers for chronic atrophic gastritis from serum exosomal microRNAs. BMC Cancer 2019; 19:129. [PMID: 30736753 PMCID: PMC6368711 DOI: 10.1186/s12885-019-5328-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 01/29/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Serum exosomal microRNAs (miRNAs) have been suggested as novel biomarkers for various diseases, especially gastric cancer (GC). But circulating biomarkers for Chronic atrophic gastritis (CAG) which is defined as precancrerous lesions of GC remain largely elusive. To investigate serum exosomal miRNAs that are differently expressed in CAG patients and Chronic nonatrophic gastritis (CNAG) may be helpful for its diagnosis and therapy. METHODS Patients were recruited according to the diagnosis and exclusioncriteria. RNA was extracted from serum exosomes of 30 CAG and 30 CNAG patients. The miRNA expression profiles were analyzed by next generation sequencing and were validated by qRT-PCR. Receiver operating characteristic (ROC) analysis has been used to evaluate the diagnostic value. RESULTS 30 CAG patients and 30 CNAG patients were recruited in our study. sRNA-seq results showed that hsa-miR-3591-3p, - 122-3p, and - 122-5p of the top 10 miRNAs (hsa-miR-148a-3p, - 122-3p, - 486-3p, -451a, - 122-5p, - 3591-3p, - 486-5p, -151a-3p, -92a-3p, -320a) were significantly upregulated in exosomes from CAG patients versus those from CNAG patients, but hsa-miR-451a, -151a-3p, and -92a-3p were significantly downregulated. Furthermore, qRT-PCR analysis confirmed that hsa-miR-122-5p and hsa-miR-122-3p were significantly upregulated in CAG samples, but hsa-miR-122-3p hadnot a steable expression. ROC curves showed that the AUC for hsa-miR-122-5p was 0.67 (95% CI 0.52-0.82, SE 62%, SP 86%). A sum of the four miRNAs (panel 1, hsa-miR-122-5p, -451a, -151a-3p, and -92a-3p) did not significantly improve the diagnostic potential (AUC 0.63, 95% CI 0.47 to 0.78). Correlation analysis showed that the expression of hsa-miR-122-5p differed significantly between patients based on atrophic (Moderate atrophic vs. Absent, P value was 0.036.) and IM (compare moderate-severe, absent and mild P values were 0.001 and 0.014, respectively). However, there were no differences between groups based on age, gender, dysplasia, or chronic or active inflammation. CONCLUSION These results suggested that hsa-miR-122-5p in serum exosomes might serve as a potential biomarker for CAG diagnosis. TRIAL REGISTRATION Chinese Clinical Trial Registy ( ChiCTR-IOR-16008027 , Date of Registration:2016-03-01).
Collapse
Affiliation(s)
- Hong Liu
- The First Affiliated Hospital of Guangdong Pharmaceutical University, No.19 Nonglinxia Road, Guangzhou, 510080 Guangdong Province China
- Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
| | - Pei-wu Li
- Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No.16 Jichang Road, Guangzhou, 510405 Guangdong Province China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
| | - Wei-qin Yang
- The Eight Affiliated Hospital, Sun Yat-sen University, No.3025 Shennanzhong Road, Shenzhen, 518033 Guangdong Province China
| | - Hong Mi
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No.16 Jichang Road, Guangzhou, 510405 Guangdong Province China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
| | - Jing-lin Pan
- Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
| | - Yuan-cheng Huang
- Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
| | - Zheng-kun Hou
- Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No.16 Jichang Road, Guangzhou, 510405 Guangdong Province China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
| | - Qiu-ke Hou
- Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No.16 Jichang Road, Guangzhou, 510405 Guangdong Province China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
| | - Qi Luo
- Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No.16 Jichang Road, Guangzhou, 510405 Guangdong Province China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
| | - Feng-bin Liu
- Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No.16 Jichang Road, Guangzhou, 510405 Guangdong Province China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Jichang Road, Guangzhou, 510405 Guangdong Province China
| |
Collapse
|
17
|
Minogue TD, Koehler JW, Stefan CP, Conrad TA. Next-Generation Sequencing for Biodefense: Biothreat Detection, Forensics, and the Clinic. Clin Chem 2018; 65:383-392. [PMID: 30352865 DOI: 10.1373/clinchem.2016.266536] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/22/2018] [Indexed: 11/06/2022]
Abstract
BACKGROUND Next-generation sequencing (NGS) is revolutionizing a variety of molecular biology fields including bioforensics, biosurveillance, and infectious disease diagnostics. For pathogen detection, the ability to sequence all nucleic acids in a sample allows near limitless multiplexability, free from a priori knowledge regarding an etiologic agent as is typically required for targeted molecular assays such as real-time PCR. Furthermore, sequencing capabilities can generate in depth genomic information, allowing detailed molecular epidemiological studies and bioforensics analysis, which is critical for source agent identification in a biothreat outbreak. However, lack of analytical specificity, inherent to NGS, presents challenges for regulated applications such as clinical diagnostics and molecular attribution. CONTENT Here, we discuss NGS applications in the context of preparedness and biothreat readiness. Specifically, we investigate current and future applications of NGS technologies to affect the fields of biosurveillance, bioforensics, and clinical diagnostics with specific focus on biodefense. SUMMARY Overall, there are many advantages to the implementation of NGS for preparedness and readiness against biowarfare agents, from forensics to diagnostics. However, appropriate caveats must be associated with any technology. This includes NGS. While NGS is not the panacea replacing all molecular techniques, it will greatly enhance the ability to detect, characterize, and diagnose biowarfare agents, thus providing an excellent addition to the biodefense toolbox of biosurveillance, bioforensics, and biothreat diagnosis.
Collapse
Affiliation(s)
- Timothy D Minogue
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD.
| | - Jeffrey W Koehler
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD
| | - Christopher P Stefan
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD
| | - Turner A Conrad
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD
| |
Collapse
|
18
|
Lee IJ, Goo NI, Kim DE. Label/quencher-free detection of single-nucleotide changes in DNA using isothermal amplification and G-quadruplexes. Analyst 2018; 141:6503-6506. [PMID: 27827492 DOI: 10.1039/c6an01600f] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We report an analytical method that exploits the interaction between G-quadruplexes and thioflavin T (ThT), for detecting mutant DNA species containing single-base changes. This system is a label/quencher-free fluorescence enhancement system based on rolling circle amplification (RCA)-responsive G-quadruplex formation, which enables a highly selective detection of T790M SNPs in the gene encoding the epidermal growth factor receptor, EGFR.
Collapse
Affiliation(s)
- Il Joon Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea.
| | - Nam-In Goo
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea.
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
19
|
Conrad TA, Lo CC, Koehler JW, Graham AS, Stefan CP, Hall AT, Douglas CE, Chain PS, Minogue TD. Diagnostic targETEd seQuencing adjudicaTion (DETEQT): Algorithms for Adjudicating Targeted Infectious Disease Next-Generation Sequencing Panels. J Mol Diagn 2018; 21:99-110. [PMID: 30268944 DOI: 10.1016/j.jmoldx.2018.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/23/2018] [Accepted: 08/14/2018] [Indexed: 12/28/2022] Open
Abstract
Next-generation sequencing (NGS) for infectious disease diagnostics is a relatively new and underdeveloped concept. If this technology is to become a regulatory-grade clinical diagnostic, standardization in the form of locked-down assays and firmly established underlying processes is necessary. Targeted sequencing, specifically by amplification of genomic signatures, has the potential to bridge the gap between PCR- and NGS-based diagnostics; however, existing NGS assay panels lack validated analytical techniques to adjudicate high background and error-prone NGS data. Herein, we present the Diagnostic targETEd seQuencing adjudicaTion (DETEQT) software, consisting of an intuitive bioinformatics pipeline entailing a set of algorithms to translate raw sequencing data into positive, negative, and indeterminate diagnostic determinations. After basic read filtering and mapping, the software compares abundance and quality metrics against heuristic and fixed thresholds. A novel generalized quality function provides an amalgamated quality score for the match between sequence reads of an assay and panel targets, rather than considering each component factor independently. When evaluated against numerous assay samples and parameters (mock clinical, human, and nonhuman primate clinical data sets; diverse amplification strategies; downstream applications; and sequence platforms), DETEQT demonstrated improved rejection of false positives and accuracies >95%. Finally, DETEQT was implemented in the user-friendly Empowering the Development of Genomics Expertise (EDGE) bioinformatics platform, providing a complete, end-to-end solution that can be operated by nonexperts in a clinical laboratory setting.
Collapse
Affiliation(s)
- Turner A Conrad
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | - Chien-Chi Lo
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico
| | - Jeffrey W Koehler
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | - Amanda S Graham
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | - Christopher P Stefan
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | - Adrienne T Hall
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | - Christina E Douglas
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | - Patrick S Chain
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico
| | - Timothy D Minogue
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland.
| |
Collapse
|
20
|
Evaluation of Targeted Next-Generation Sequencing for Detection of Bovine Pathogens in Clinical Samples. J Clin Microbiol 2018; 56:JCM.00399-18. [PMID: 29695524 PMCID: PMC6018347 DOI: 10.1128/jcm.00399-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/18/2018] [Indexed: 02/07/2023] Open
Abstract
The laboratory diagnosis of infectious diseases, especially those caused by mixed infections, is challenging. Routinely, it requires submission of multiple samples to separate laboratories. Advances in next-generation sequencing (NGS) have provided the opportunity for development of a comprehensive method to identify infectious agents. This study describes the use of target-specific primers for PCR-mediated amplification with the NGS technology in which pathogen genomic regions of interest are enriched and selectively sequenced from clinical samples. In the study, 198 primers were designed to target 43 common bovine and small-ruminant bacterial, fungal, viral, and parasitic pathogens, and a bioinformatics tool was specifically constructed for the detection of targeted pathogens. The primers were confirmed to detect the intended pathogens by testing reference strains and isolates. The method was then validated using 60 clinical samples (including tissues, feces, and milk) that were also tested with other routine diagnostic techniques. The detection limits of the targeted NGS method were evaluated using 10 representative pathogens that were also tested by quantitative PCR (qPCR), and the NGS method was able to detect the organisms from samples with qPCR threshold cycle (CT) values in the 30s. The method was successful for the detection of multiple pathogens in the clinical samples, including some additional pathogens missed by the routine techniques because the specific tests needed for the particular organisms were not performed. The results demonstrate the feasibility of the approach and indicate that it is possible to incorporate NGS as a diagnostic tool in a cost-effective manner into a veterinary diagnostic laboratory.
Collapse
|
21
|
Shinozuka H, Sudheesh S, Shinozuka M, Cogan NOI. Homology-based enzymatic DNA fragment assembly-based illumina sequencing library preparation. Biol Methods Protoc 2018; 3:bpy001. [PMID: 32161795 PMCID: PMC6994068 DOI: 10.1093/biomethods/bpy001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/03/2018] [Accepted: 01/19/2018] [Indexed: 11/14/2022] Open
Abstract
The current Illumina HiSeq and MiSeq platforms can generate paired-end reads of up to 2 x 250 bp and 2 x 300 bp in length, respectively. These read lengths may be substantially longer than genomic regions of interest when a DNA sequencing library is prepared through a target enrichment-based approach. A sequencing library preparation method has been developed based on the homology-based enzymatic DNA fragment assembly scheme to allow processing of multiple PCR products within a single read. Target sequences were amplified using locus-specific PCR primers with 8 bp tags, and using the tags, homology-based enzymatic DNA assembly was performed with DNA polymerase, T7 exonuclease and T4 DNA ligase. Short PCR amplicons can hence be assembled into a single molecule, along with sequencing adapters specific to the Illumina platforms. As a proof-of-concept experiment, short PCR amplicons (57-66 bp in length) derived from genomic DNA templates of field pea and containing variable nucleotide locations were assembled and sequenced on the MiSeq platform. The results were validated with other genotyping methods. When 5 PCR amplicons were assembled, 4.3 targeted sequences (single-nucleotide polymorphisms) on average were successfully identified within each read. The utility of this for sequencing of short fragments has consequently been demonstrated.
Collapse
Affiliation(s)
- Hiroshi Shinozuka
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, 5 Ring Road, La Trobe University, Bundoora, Victoria, 3083
| | - Shimna Sudheesh
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, 5 Ring Road, La Trobe University, Bundoora, Victoria, 3083
| | - Maiko Shinozuka
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, 5 Ring Road, La Trobe University, Bundoora, Victoria, 3083
| | - Noel O I Cogan
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, 5 Ring Road, La Trobe University, Bundoora, Victoria, 3083
- School of Applied Systems Biology, La Trobe University, Bundoora, Victoria, 3086, Australia
| |
Collapse
|
22
|
Detection of 16S rRNA and KPC Genes from Complex Matrix Utilizing a Molecular Inversion Probe Assay for Next-Generation Sequencing. Sci Rep 2018; 8:2028. [PMID: 29391471 PMCID: PMC5794912 DOI: 10.1038/s41598-018-19501-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 12/27/2017] [Indexed: 11/08/2022] Open
Abstract
Targeted sequencing promises to bring next-generation sequencing (NGS) into routine clinical use for infectious disease diagnostics. In this context, upfront processing techniques, including pathogen signature enrichment, must amplify multiple targets of interest for NGS to be relevant when applied to patient samples with limited volumes. Here, we demonstrate an optimized molecular inversion probe (MIP) assay targeting multiple variable regions within the 16S ribosomal gene for the identification of biothreat and ESKAPE pathogens in a process that significantly reduces complexity, labor, and processing time. Probes targeting the Klebsiella pneumoniae carbapenemase (KPC) antibiotic resistance (AR) gene were also included to demonstrate the ability to concurrently identify etiologic agent and ascertain valuable secondary genetic information. Our assay captured gene sequences in 100% of mock clinical samples prepared from flagged positive blood culture bottles. Using a simplified processing and adjudication method for mapped sequencing reads, genus and species level concordance was 100% and 80%, respectively. In addition, sensitivity and specificity for KPC gene detection was 100%. Our MIP assay produced sequenceable amplicons for the identification of etiologic agents and the detection of AR genes directly from blood culture bottles in a simplified single tube assay.
Collapse
|
23
|
Sharma D, Patel RP, Zaidi STR, Sarker MMR, Lean QY, Ming LC. Interplay of the Quality of Ciprofloxacin and Antibiotic Resistance in Developing Countries. Front Pharmacol 2017; 8:546. [PMID: 28871228 PMCID: PMC5566961 DOI: 10.3389/fphar.2017.00546] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/04/2017] [Indexed: 12/01/2022] Open
Abstract
Ciprofloxacin, a second generation broad spectrum fluoroquinolone, is active against both Gram-positive and Gram-negative bacteria. Ciprofloxacin has a high oral bioavailability and a large volume of distribution. It is used for the treatment of a wide range of infections including urinary tract infections caused by susceptible bacteria. However, the availability and use of substandard and spurious quality of oral ciprofloxacin formulations in the developing countries has been thought to have contributed toward increased risk of treatment failure and bacterial resistance. Therefore, quality control and bioequivalence studies of the commercially available oral ciprofloxacin formulations should be monitored. Appropriate actions should be taken against offending manufacturers in order to prevent the sale of substandard and spurious quality of ciprofloxacin formulations.
Collapse
Affiliation(s)
- Deepali Sharma
- Pharmacy, School of Medicine, University of Tasmania, HobartTAS, Australia
| | - Rahul P Patel
- Pharmacy, School of Medicine, University of Tasmania, HobartTAS, Australia
| | | | | | - Qi Ying Lean
- Vector borne Diseases Research Group, Pharmaceutical and Life Sciences CoRe, Universiti Teknologi MARAShah Alam, Malaysia.,Faculty of Pharmacy, Universiti Teknologi MARABertam, Malaysia
| | - Long C Ming
- Pharmacy, School of Medicine, University of Tasmania, HobartTAS, Australia.,School of Pharmacy, KPJ Healthcare University CollegeNegeri Sembilan, Malaysia
| |
Collapse
|
24
|
Abstract
Circulating exosome holds great potentials as biomarker for diagnosis and prognosis of human cancers. Previously, we have applied small RNA sequencing to identify aberrantly expressed exosomal miRNAs as candidates for diagnostic markers in colon cancer patients. In this validation cohort, plasma derived exosomal miRNA was isolated from 50 early-stage colon cancer patients and 50 matched healthy volunteers. Real-time qRT-PCR revealed that miR-125a-3p, miR-320c were significantly up-regulated in plasma exosomes of the patients with early stage colon cancer. ROC curve showed that miR-125a-3p abundant level may predict colon cancer with an area of under the curve (AUC) of 68.5%, in comparison to that of CEA at 83.6%. Combination of miR-125a-3P and CEA improved the AUC to 85.5%. In addition, plasma exosome level of miR-125a-3p and miR-320c showed significant correlation with nerve infiltration (P < 0.01), but not with tumor size, infiltration depth, and differentiation degree (P > 0.05). On the contrary, plasma CEA level is correlated with tumor size, infiltration depth, and differentiation degree (P < 0.05, r = 0.3009–0.7270), but not with nerve infiltration (P = 0.744). In conclusion, this follow-up study demonstrated circulating plasma exosomal miR-125a-3p is readily accessible as diagnosis biomarker for early-stage colon cancer. When combined with conventional diagnostic markers, miR-125a-3p can improve the diagnostic power.
Collapse
|