1
|
Yoshii K, Node E, Furuta M, Tojima Y, Matsunaga A, Adachi J, Takaai N, Morita M, Hosomi K, Kunisawa J. Establishment of enterotype-specific antibodies for various diagnostic systems. Sci Rep 2025; 15:16814. [PMID: 40368953 PMCID: PMC12078515 DOI: 10.1038/s41598-025-01144-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 05/05/2025] [Indexed: 05/16/2025] Open
Abstract
This study demonstrates that monoclonal antibodies can be developed to targeting specific gut bacteria prevalent in the Japanese population and the potential for creating a novel diagnostic system using these antibodies. In this study, we established specific antibodies against representative bacteria from the genera Bacteroides, Faecalibacterium, and Prevotella and showed that they could be detected using ELISA, flow cytometry, and western blot analysis. Furthermore, a technique to quantify target bacteria was developed by combining these antibodies in a sandwich ELISA, enabling the quantification of bacteria in human fecal samples. This technology serves as a foundational method for rapidly and easily measuring gut bacteria and is expected to evolve into a powerful tool for analyzing the impact of gut bacteria on health, as well as for personalized health management based on individual gut environments.
Collapse
Affiliation(s)
- Ken Yoshii
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Eri Node
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Mari Furuta
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Yoko Tojima
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Ayu Matsunaga
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
- Faculty of Agriculture, Department of Applied Biological Science, Takasaki University of Health and Welfare, 54 Nakaorui-machi, Takasaki, Gunma, 370-0033, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, NIBN, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Narimi Takaai
- Laboratory of Proteomics for Drug Discovery, NIBN, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Makiko Morita
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
- Graduate School of Veterinary Science, Osaka Metropolitan University, 1-58 Rinku-oraikita, Izumisano, Osaka, 598-8531, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan.
- Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Graduate School of Science, Osaka University, 1-1 Machikaneyamacho, Toyonaka, Osaka, 560-0043, Japan.
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
- Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
- Research Organization for Nano and Life Innovation, Waseda University, 2-2 Wakamatsu, Shinjuku-ku, Tokyo, 162-8480, Japan.
| |
Collapse
|
2
|
Rapszky GA, Do To UN, Kiss VE, Kói T, Walter A, Gergő D, Meznerics FA, Rakovics M, Váncsa S, Kemény LV, Csupor D, Hegyi P, Filbin MR, Varga C, Fenyves BG. Rapid molecular assays versus blood culture for bloodstream infections: a systematic review and meta-analysis. EClinicalMedicine 2025; 79:103028. [PMID: 39968206 PMCID: PMC11833021 DOI: 10.1016/j.eclinm.2024.103028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 02/20/2025] Open
Abstract
Background Timely management of sepsis with early targeted antimicrobial therapy improves patient outcomes. Rapid molecular assays (RMAs) have emerged, enabling the detection of bloodstream infection (BSI) with a shorter turnaround time than blood cultures (BCs). The accuracy of several RMAs has not been comprehensively reviewed. We aimed to identify commercial RMAs reported in the literature and evaluate their diagnostic performance compared to BC. Methods A systematic review and meta-analysis was conducted, covering MEDLINE, Cochrane Library, Embase, and Web of Science from inception to September 23, 2024. Eligible studies included patients with suspected or documented BSI, tested with both an RMA (turnaround time of ≤12 h, targeting ≥20 pathogens) and BC. Non-original research articles and animal studies were excluded. The primary outcomes were pooled sensitivity and specificity of RMAs for pathogen detection compared to BC. Bivariate analysis was used to produce summary receiver operating characteristic plots and diagnostic metric measures stratified by different units of analysis (sample versus patient), RMA types, and patient populations. Risk of bias was assessed using the Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) and Quality Assessment of Diagnostic Accuracy Studies-Comparative (QUADAS-C) tools. The study was registered with PROSPERO, CRD42022377280. Findings A total of 63,916 articles were identified, of which 104 were included in the qualitative synthesis and 75 in the quantitative synthesis, covering 17,952 samples and 11,393 patients analyzed separately. Eleven RMAs were identified, with four included in the RMA-based subgroup analysis (LightCycler SeptiFast Test MGRADE®, IRIDICA BAC BSI assay, SepsiTest, MagicPlex Sepsis Test) and five additional ones in the pooled analysis (UMD-SelectNA, VYOO®, MicrobScan assay, MicrobScan-Kairos24/7, REBA Sepsis-ID test). Two RMAs were included in the qualitative synthesis only (InfectID-BSI, Pilot Gene Technology droplet digital polymerase chain reaction). Pooled specificity of RMAs was higher (0.858, 95% confidence interval (CI) 0.830-0.883) than sensitivity (0.659, 95% CI 0.594-0.719) by patient. Sensitivities varied by RMA type from 0.492 (95% CI 0.390-0.594, MagicPlex Sepsis Test) to 0.783 (95% CI 0.662-0.870, IRIDICA BAC BSI assay) by patient. Specificities varied more by patient population, ranging from 0.811 (95% CI 0.716-0.879) in the intensive care population to 0.892 (95% CI 0.838-0.930) in the emergency department population, by patient. Similar metrics were observed when the analysis was done by sample. Risk of bias was judged to be high in all included articles. Interpretation Despite their shorter turnaround time, low sensitivity means RMAs cannot replace BCs. However, our data indicate that RMAs may have value as an add-on test by increasing pathogen detection rates. Higher-sensitivity RMAs are needed which could possibly be achieved by expanding pathogen coverage and increasing blood sample volumes. High-quality implementation studies and standardized reporting are required to assess the clinical advantages of RMAs. Funding Centre for Translational Medicine, Semmelweis University.
Collapse
Affiliation(s)
- Gabriella Anna Rapszky
- Department of Emergency Medicine, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Uyen Nguyen Do To
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- András Pető Faculty, Semmelweis University, Budapest, Hungary
| | | | - Tamás Kói
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Budapest University of Technology and Economics, Department of Stochastics, Budapest, Hungary
| | - Anna Walter
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Dorottya Gergő
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Pharmacognosy, Semmelweis University, Budapest, Hungary
| | - Fanni Adél Meznerics
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - Márton Rakovics
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Eötvös Loránd University, Faculty of Social Sciences, Department of Statistics, Budapest, Hungary
| | - Szilárd Váncsa
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
| | - Lajos Vince Kemény
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
- Department of Physiology, Semmelweis University, Budapest, Hungary
- HCEMM-SU, Translational Dermatology Research Group, Semmelweis University, Budapest, Hungary
| | - Dezső Csupor
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Clinical Pharmacy, University of Szeged, Szeged, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
| | - Michael R. Filbin
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Emergency Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Csaba Varga
- Department of Emergency Medicine, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Bánk G. Fenyves
- Department of Emergency Medicine, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
3
|
Zhang J, He Y, Chen C, Hu W, He J, Ying Y, Zhu F. Bacterial Analysis of the Whole Blood in Chinese Healthy Donors Using 16S rDNA-Targeted Metagenomic Sequencing. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:6635560. [PMID: 39444936 PMCID: PMC11498981 DOI: 10.1155/2024/6635560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Background: The presence of bacteria in the blood of healthy individuals remains controversial. This study explored the comprehensive bacterial profiles and specific biomarkers in different components of healthy Chinese blood donors. Methods: A total of 5230 whole blood (WB) specimens were collected. Among them, 5200 random samples were pooled into 26 mixed samples for bacterial profile analysis. The remaining 30 random samples were divided into 4 groups based on components: WB, plasma, red blood cells (RBCs), and buffy coat (BC). Subsequently, the amplicons of the bacterial 16S rDNA V3-V4 fragments were sequenced to measure the diversity and composition of the bacteria using next-generation sequencing. Results: The bacterial DNAs in the blood primarily originated from the Proteobacteria phylum. A total of 301 species of bacterial DNA were found in blood specimens, with 46 species being present among all groups. A significantly higher abundance of bacterial DNA was found in the plasma and RBCs compared to those in BC and WB. However, the plasma and RBC groups showed significantly higher species diversity and richness compared to the BC and WB groups. In addition, the WB group had a significantly different community structure and composition compared to the plasma and RBC groups but was similar to the BC group. Conclusion: The presence of bacterial DNA fragments was confirmed in blood from healthy Chinese donors. The bacterial DNA fragments enriched in plasma showed the highest diversity, followed by RBC, WB, and BC. These results provide a foundation for further research on the microbiome in the blood of healthy individuals.
Collapse
Affiliation(s)
- Jingjing Zhang
- Institute of Transfusion Medicine, Blood Center of Zhejiang Province, Hangzhou, China
| | - Yanmin He
- Institute of Transfusion Medicine, Blood Center of Zhejiang Province, Hangzhou, China
| | - Chen Chen
- Institute of Transfusion Medicine, Blood Center of Zhejiang Province, Hangzhou, China
| | - Wei Hu
- Institute of Transfusion Medicine, Blood Center of Zhejiang Province, Hangzhou, China
| | - Ji He
- Institute of Transfusion Medicine, Blood Center of Zhejiang Province, Hangzhou, China
| | - Yanling Ying
- Institute of Transfusion Medicine, Blood Center of Zhejiang Province, Hangzhou, China
| | - Faming Zhu
- Institute of Transfusion Medicine, Blood Center of Zhejiang Province, Hangzhou, China
| |
Collapse
|
4
|
Jones KL, Cunha F, Casaro S, Galvão KN. Optimization and Testing of a Commercial Viability PCR Protocol to Detect Escherichia coli in Whole Blood. Microorganisms 2024; 12:765. [PMID: 38674709 PMCID: PMC11052410 DOI: 10.3390/microorganisms12040765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Bacteremia, specifically if progressed to sepsis, poses a time-sensitive threat to human and animal health. Escherichia coli is a main causative agent of sepsis in humans. The objective was to evaluate a propidium monoazide (PMA)-based viability PCR (vPCR) protocol to detect and quantify live E. coli from whole blood. We optimized the protocol by adding a eukaryotic-specific lysis step prior to PMA exposure, then used spiking experiments to determine the lower limit of detection (LOD) and linear range of quantification. We also compared the vPCR quantification method to standard colony count of spiked inoculum. Lastly, we calculated percent viability in spiked samples containing 50% live cells or 0% live cells. The LOD was 102 CFU/mL for samples containing live cells only and samples with mixed live and heat-killed cells. The linear range of quantification was 102 CFU/mL to 108 CFU/mL (R2 of 0.997) in samples containing only live cells and 103 CFU/mL to 108 CFU/mL (R2 of 0.998) in samples containing live plus heat-killed cells. A Bland-Altman analysis showed that vPCR quantification overestimates compared to standard plate count of the spiked inoculum, with an average bias of 1.85 Log10 CFU/mL across the linear range when only live cells were present in the sample and 1.98 Log10 CFU/mL when live plus heat-killed cells were present. Lastly, percent viability calculations showed an average 89.5% viable cells for samples containing 50% live cells and an average 19.3% for samples containing 0% live cells. In summary, this optimized protocol can detect and quantify viable E. coli in blood in the presence of heat-killed cells. Additionally, the data presented here provide the groundwork for further development of vPCR to detect and quantify live bacteria in blood in clinical settings.
Collapse
Affiliation(s)
| | | | | | - Klibs N. Galvão
- Department of Large Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32608, USA
| |
Collapse
|
5
|
Gieroń M, Żarnowiec P, Zegadło K, Gmiter D, Czerwonka G, Kaca W, Kręcisz B. Loop-Mediated Isothermal Amplification of DNA (LAMP) as an Alternative Method for Determining Bacteria in Wound Infections. Int J Mol Sci 2023; 25:411. [PMID: 38203582 PMCID: PMC10778741 DOI: 10.3390/ijms25010411] [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: 11/04/2023] [Revised: 12/23/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
The increasing number of patients with chronic wounds requires the development of quick and accurate diagnostics methods. One of the key and challenging aspects of treating ulcers is to control wound infection. Early detection of infection is essential for the application of suitable treatment methods, such as systemic antibiotics or other antimicrobial agents. Clinically, the most frequently used method for detecting microorganisms in wounds is through a swab and culture on appropriate media. This test has major limitations, such as the long bacterial growth time and the selectivity of bacterial growth. This article presents an overview of molecular methods for detecting bacteria in wounds, including real-time polymerase chain reaction (rtPCR), quantitative polymerase chain reaction (qPCR), genotyping, next-generation sequencing (NGS), and loop-mediated isothermal amplification (LAMP). We focus on the LAMP method, which has not yet been widely used to detect bacteria in wounds, but it is an interesting alternative to conventional detection methods. LAMP does not require additional complicated equipment and provides the fastest detection time for microorganisms (approx. 30 min reaction). It also allows the use of many pairs of primers in one reaction and determination of up to 15 organisms in one sample. Isothermal amplification of DNA is currently the easiest and most economical method for microbial detection in wound infection. Direct visualization of the reaction with dyes, along with omitting DNA isolation, has increased the potential use of this method.
Collapse
Affiliation(s)
- Monika Gieroń
- Faculty of Medicine, Jan Kochanowski University in Kielce, 25-369 Kielce, Poland; (M.G.); (B.K.)
- Dermatology Department, Provincial General Hospital, 25-317 Kielce, Poland
| | - Paulina Żarnowiec
- Department of Microbiology, Institute of Biology, Jan Kochanowski University in Kielce, 25-406 Kielce, Poland; (P.Ż.); (K.Z.); (D.G.); (W.K.)
| | - Katarzyna Zegadło
- Department of Microbiology, Institute of Biology, Jan Kochanowski University in Kielce, 25-406 Kielce, Poland; (P.Ż.); (K.Z.); (D.G.); (W.K.)
| | - Dawid Gmiter
- Department of Microbiology, Institute of Biology, Jan Kochanowski University in Kielce, 25-406 Kielce, Poland; (P.Ż.); (K.Z.); (D.G.); (W.K.)
| | - Grzegorz Czerwonka
- Department of Microbiology, Institute of Biology, Jan Kochanowski University in Kielce, 25-406 Kielce, Poland; (P.Ż.); (K.Z.); (D.G.); (W.K.)
| | - Wiesław Kaca
- Department of Microbiology, Institute of Biology, Jan Kochanowski University in Kielce, 25-406 Kielce, Poland; (P.Ż.); (K.Z.); (D.G.); (W.K.)
| | - Beata Kręcisz
- Faculty of Medicine, Jan Kochanowski University in Kielce, 25-369 Kielce, Poland; (M.G.); (B.K.)
- Dermatology Department, Provincial General Hospital, 25-317 Kielce, Poland
| |
Collapse
|
6
|
Klein EJ, Almaghlouth NK, Weigel G, Farmakiotis D, Hardy E. Refractory Bilateral Tubo-Ovarian Abscesses in a Patient with Iatrogenic Hypogammaglobulinemia. Diagnostics (Basel) 2023; 13:3478. [PMID: 37998614 PMCID: PMC10670296 DOI: 10.3390/diagnostics13223478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/31/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023] Open
Abstract
Genital mycoplasmas are sexually transmitted Mollicutes with a high prevalence of urogenital tract colonization among females of reproductive age. Current guidelines recommend against routine screening for these organisms, since their role in the pathogenesis of pelvic inflammatory disease and tubo-ovarian abscesses (TOAs) remains unclear. However, genital mycoplasmas harbor pathogenic potential in immunocompromised hosts, especially patients with hypogammaglobulinemia. It is important to identify such infections early, given their potential for invasive spread and the availability of easily accessible treatments. We present a young adult female with multiple sclerosis and iatrogenic hypogammaglobulinemia, with refractory, bilateral pelvic inflammatory disease and TOAs due to Ureaplasma urealyticum, identified as a single pathogen via three distinct molecular tests. To our knowledge, this is the second case of TOAs caused by U. urealyticum in the literature, and the first diagnosed by pathogen cell-free DNA metagenomic next-generation sequencing in plasma.
Collapse
Affiliation(s)
- Elizabeth J. Klein
- Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA; (E.J.K.); (N.K.A.); (D.F.)
| | - Nouf K. Almaghlouth
- Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA; (E.J.K.); (N.K.A.); (D.F.)
| | - Gabriela Weigel
- Department of Obstetrics and Gynecology, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Dimitrios Farmakiotis
- Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA; (E.J.K.); (N.K.A.); (D.F.)
| | - Erica Hardy
- Divisions of Obstetric Medicine and Infectious Diseases, Women and Infants Hospital, Providence, RI 02905, USA
| |
Collapse
|
7
|
Zhang N, Li J, Zhan Y, Wang K, Zhan Z, Wei H, Zhang Z. Acid-tolerant Lactiplantibacillus plantarum ZDY2013 shows a colonization niche preference and interacts with enterotoxigenic Bacillus cereus in specific-pathogen-free mice. Food Funct 2023; 14:6410-6421. [PMID: 37366339 DOI: 10.1039/d3fo01468a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Probiotics have long been utilized as functional food and modulate gut microbial homeostasis, but their colonization niche is mostly unclear and transient, which restrains the development of microbiome-targeted strategies. Lactiplantibacillus (L.) plantarum ZDY2013 is an allochthonous species of the human gastrointestinal tract with acid-tolerant properties. It serves as an antagonistic agent against the food-borne pathogen Bacillus (B.) cereus and a potent regulator of the gut microbiota. However, there is a knowledge gap regarding the colonization dynamics of L. plantarum ZDY2013 in the host intestine and the colonization niche of its interaction with pathogens. Here, we designed a pair of specific primers targeting L. plantarum ZDY2013 based on its whole genome sequence. We evaluated their accuracy and sensitivity with other host-derived strains and confirmed their availability with artificially spiked fecal samples from different mouse models. Additionally, the content of L. plantarum ZDY2013 was quantified by qPCR in fecal samples from BALB/c mice, followed by the analysis of its colonization niche preference. Moreover, the interactions between L. plantarum ZDY2013 and enterotoxigenic B. cereus HN001 were also elucidated. The results revealed that the newly designed primers could identify L. plantarum ZDY2013 with high specificity and were resistant to the influence of the complex fecal matrix and gut microbes from different hosts. Interestingly, the content of mixed L. plantarum ZDY2013 and B. cereus HN001 when orally administered remained higher when compared with the single strain group in BALB/c mice upon discontinuation of intragastric administration. In addition, L. plantarum ZDY2013 was mainly enriched in the large intestine during the ingestion period and maintained the highest content in the stomach after discontinuing supplementation on day 7. Moreover, L. plantarum ZDY2013 colonization neither damaged the intestine nor ameliorated the damage triggered by B. cereus in BALB/c mice. Overall, our study constructed two efficient specific primers targeting L. plantarum ZDY2013 and provided the potential to explore the underlying mechanism of competition between L. plantarum ZDY2013 and pathogens in host species.
Collapse
Affiliation(s)
- Na Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Jinmei Li
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Ying Zhan
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Kaiming Wang
- Department of Physiology, CEGIIR, University of Alberta, Edmonton T6G 2E1, Canada
| | - Zhongxu Zhan
- Jiangxi General Institute of Testing and Certification Food Testing Institute, Nanchang, 330200, China
| | - Hua Wei
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China
| | - Zhihong Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
- International Institute of Food Innovation, Nanchang University, Nanchang, 330299, China
| |
Collapse
|
8
|
Marutescu LG. Current and Future Flow Cytometry Applications Contributing to Antimicrobial Resistance Control. Microorganisms 2023; 11:1300. [PMID: 37317273 DOI: 10.3390/microorganisms11051300] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 06/16/2023] Open
Abstract
Antimicrobial resistance is a global threat to human health and welfare, food safety, and environmental health. The rapid detection and quantification of antimicrobial resistance are important for both infectious disease control and public health threat assessment. Technologies such as flow cytometry can provide clinicians with the early information, they need for appropriate antibiotic treatment. At the same time, cytometry platforms facilitate the measurement of antibiotic-resistant bacteria in environments impacted by human activities, enabling assessment of their impact on watersheds and soils. This review focuses on the latest applications of flow cytometry for the detection of pathogens and antibiotic-resistant bacteria in both clinical and environmental samples. Novel antimicrobial susceptibility testing frameworks embedding flow cytometry assays can contribute to the implementation of global antimicrobial resistance surveillance systems that are needed for science-based decisions and actions.
Collapse
Affiliation(s)
- Luminita Gabriela Marutescu
- Department of Botany and Microbiology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania
- Research Institute of the University of Bucharest, 050095 Bucharest, Romania
| |
Collapse
|
9
|
Scheuermann-Poley C, Wiessner A, Kikhney J, Gatzer R, Müller M, Stichling M, Moter A, Willy C. Fluorescence In Situ Hybridization as Diagnostic Tool for Implant-associated Infections: A Pilot Study on Added Value. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2023; 11:e4994. [PMID: 37360245 PMCID: PMC10287136 DOI: 10.1097/gox.0000000000004994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 03/23/2023] [Indexed: 06/28/2023]
Abstract
Implant-associated infections are a devastating complication in surgery. Especially in infections with biofilm-forming microorganisms, the identification of the causing microorganism remains a challenge. However, the classification as biofilm is not possible with conventional polymerase chain reaction or culture-based diagnostics. The aim of this study was to evaluate the additional value of fluorescence in situ hybridization (FISH) and nucleic acid amplification technique (FISHseq) to discuss a diagnostic benefit of the culture-independent methods and to map spatial organization of pathogens and microbial biofilms in wounds. Methods In total, 118 tissue samples from 60 patients with clinically suspected implant-associated infections (n = 32 joint replacements, n = 24 open reduction and internal fixation, n = 4 projectiles) were analyzed using classic microbiological culture and culture-independent FISH in combination with polymerase chain reaction and sequencing (FISHseq). Results In 56 of 60 wounds, FISHseq achieved an added value. FISHseq confirmed the result of cultural microbiological examinations in 41 of the 60 wounds. In 12 wounds, one or more additional pathogens were detected by FISHseq. FISHseq could show that the bacteria initially detected by culture corresponded to a contamination in three wounds and could exclude that the identified commensal pathogens were a contamination in four other wounds. In five wounds, a nonplanktonic bacterial life form was detected. Conclusions The study revealed that FISHseq gives additional diagnostic information, including therapy-relevant findings that were missed by culture. In addition, nonplanktonic bacterial life forms could also be detected with FISHseq, albeit less frequently than previously indicated.
Collapse
Affiliation(s)
- Catharina Scheuermann-Poley
- From the Trauma & Orthopedic Surgery, Septic & Reconstructive Surgery, Research and Treatment Centre Septic Defect Wounds, Federal Armed Forces of Germany, Bundeswehr (Military) Academic Hospital, Berlin, Germany
| | - Alexandra Wiessner
- Biofilmcenter, Institute for Microbiology, Infectious Diseases, and Immunology, Charité – University Medicine Berlin and MoKi Analytics GmbH, Berlin, Germany
| | - Judith Kikhney
- Biofilmcenter, Institute for Microbiology, Infectious Diseases, and Immunology, Charité – University Medicine Berlin and MoKi Analytics GmbH, Berlin, Germany
| | - Renate Gatzer
- Department of Microbiology, Federal Armed Forces of Germany, Bundeswehr (Military) Academic Hospital, Berlin, Germany
| | - Martin Müller
- Department of Microbiology, Federal Armed Forces of Germany, Bundeswehr (Military) Academic Hospital, Berlin, Germany
| | - Marcus Stichling
- From the Trauma & Orthopedic Surgery, Septic & Reconstructive Surgery, Research and Treatment Centre Septic Defect Wounds, Federal Armed Forces of Germany, Bundeswehr (Military) Academic Hospital, Berlin, Germany
| | - Annette Moter
- Biofilmcenter, Institute for Microbiology, Infectious Diseases, and Immunology, Charité – University Medicine Berlin and MoKi Analytics GmbH, Berlin, Germany
| | - Christian Willy
- From the Trauma & Orthopedic Surgery, Septic & Reconstructive Surgery, Research and Treatment Centre Septic Defect Wounds, Federal Armed Forces of Germany, Bundeswehr (Military) Academic Hospital, Berlin, Germany
| |
Collapse
|
10
|
Molecular Methods for Pathogenic Bacteria Detection and Recent Advances in Wastewater Analysis. WATER 2021. [DOI: 10.3390/w13243551] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
With increasing concerns about public health and the development of molecular techniques, new detection tools and the combination of existing approaches have increased the abilities of pathogenic bacteria monitoring by exploring new biomarkers, increasing the sensitivity and accuracy of detection, quantification, and analyzing various genes such as functional genes and antimicrobial resistance genes (ARG). Molecular methods are gradually emerging as the most popular detection approach for pathogens, in addition to the conventional culture-based plate enumeration methods. The analysis of pathogens in wastewater and the back-estimation of infections in the community, also known as wastewater-based epidemiology (WBE), is an emerging methodology and has a great potential to supplement current surveillance systems for the monitoring of infectious diseases and the early warning of outbreaks. However, as a complex matrix, wastewater largely challenges the analytical performance of molecular methods. This review synthesized the literature of typical pathogenic bacteria in wastewater, types of biomarkers, molecular methods for bacterial analysis, and their recent advances in wastewater analysis. The advantages and limitation of these molecular methods were evaluated, and their prospects in WBE were discussed to provide insight for future development.
Collapse
|
11
|
Wilson-Nieuwenhuis J, El-Mohtadi M, Edwards K, Whitehead K, Dempsey-Hibbert N. Factors Involved in the onset of infection following bacterially contaminated platelet transfusions. Platelets 2021; 32:909-918. [PMID: 32762589 DOI: 10.1080/09537104.2020.1803253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Transfusion of platelet concentrates (PCs) is associated with several adverse patient reactions, the most common of which are febrile non-hemolytic transfusion reactions (FNHTRs) and transfusion-associated bacterial-infection/transfusion-associated sepsis (T-ABI/TA-S). Diagnosis of T-ABI/T-AS requires a positive blood culture (BC) result from the transfusion recipient and also a positive identification of bacterial contamination within a test aliquot of the transfused PC. In a significant number of cases, clinical symptoms post-transfusion are reported by the clinician, yet the BCs from the patient and/or PC are negative. The topic of 'missed bacterial detection' has therefore been the focus of several primary research studies and review articles, suggesting that biofilm formation in the blood bag and the presence of viable but non-culturable (VBNC) pathogens are the major causes of this missed detection. However, platelets are emerging as key players in early host responses to infection and as such, the aforementioned biofilm formation could elicit 'platelet priming', which could lead to significant immunological reactions in the host, in the absence of planktonic bacteria in the host bloodstream. This review reflects on what is known about missed detection and relates this to the emerging understanding of the effect of bacterial contamination on the platelets themselves and the significant role played by platelets in exacerbation of an immune response to infection within the transfusion setting.
Collapse
Affiliation(s)
| | - Mohamed El-Mohtadi
- Centre for Bioscience, Manchester Metropolitan University, Manchester, UK
| | - Kurtis Edwards
- Centre for Bioscience, Manchester Metropolitan University, Manchester, UK
| | - Kathryn Whitehead
- Centre for Bioscience, Manchester Metropolitan University, Manchester, UK
| | | |
Collapse
|
12
|
Kondori N, Kurtovic A, Piñeiro-Iglesias B, Salvà-Serra F, Jaén-Luchoro D, Andersson B, Alves G, Ogurtsov A, Thorsell A, Fuchs J, Tunovic T, Kamenska N, Karlsson A, Yu YK, Moore ERB, Karlsson R. Mass Spectrometry Proteotyping-Based Detection and Identification of Staphylococcus aureus, Escherichia coli, and Candida albicans in Blood. Front Cell Infect Microbiol 2021; 11:634215. [PMID: 34381737 PMCID: PMC8350517 DOI: 10.3389/fcimb.2021.634215] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Bloodstream infections (BSIs), the presence of microorganisms in blood, are potentially serious conditions that can quickly develop into sepsis and life-threatening situations. When assessing proper treatment, rapid diagnosis is the key; besides clinical judgement performed by attending physicians, supporting microbiological tests typically are performed, often requiring microbial isolation and culturing steps, which increases the time required for confirming positive cases of BSI. The additional waiting time forces physicians to prescribe broad-spectrum antibiotics and empirically based treatments, before determining the precise cause of the disease. Thus, alternative and more rapid cultivation-independent methods are needed to improve clinical diagnostics, supporting prompt and accurate treatment and reducing the development of antibiotic resistance. In this study, a culture-independent workflow for pathogen detection and identification in blood samples was developed, using peptide biomarkers and applying bottom-up proteomics analyses, i.e., so-called "proteotyping". To demonstrate the feasibility of detection of blood infectious pathogens, using proteotyping, Escherichia coli and Staphylococcus aureus were included in the study, as the most prominent bacterial causes of bacteremia and sepsis, as well as Candida albicans, one of the most prominent causes of fungemia. Model systems including spiked negative blood samples, as well as positive blood cultures, without further culturing steps, were investigated. Furthermore, an experiment designed to determine the incubation time needed for correct identification of the infectious pathogens in blood cultures was performed. The results for the spiked negative blood samples showed that proteotyping was 100- to 1,000-fold more sensitive, in comparison with the MALDI-TOF MS-based approach. Furthermore, in the analyses of ten positive blood cultures each of E. coli and S. aureus, both the MALDI-TOF MS-based and proteotyping approaches were successful in the identification of E. coli, although only proteotyping could identify S. aureus correctly in all samples. Compared with the MALDI-TOF MS-based approaches, shotgun proteotyping demonstrated higher sensitivity and accuracy, and required significantly shorter incubation time before detection and identification of the correct pathogen could be accomplished.
Collapse
Affiliation(s)
- Nahid Kondori
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Amra Kurtovic
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Francisco Salvà-Serra
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Culture Collection University of Gothenburg (CCUG), Sahlgrenska Academy of the University of Gothenburg, Gothenburg, Sweden
- Microbiology, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain
| | - Daniel Jaén-Luchoro
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Culture Collection University of Gothenburg (CCUG), Sahlgrenska Academy of the University of Gothenburg, Gothenburg, Sweden
| | - Björn Andersson
- Bioinformatics Core Facility at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Gelio Alves
- National Center for Biotechnology Information (NCBI), Bethesda, MD, United States
| | - Aleksey Ogurtsov
- National Center for Biotechnology Information (NCBI), Bethesda, MD, United States
| | - Annika Thorsell
- Proteomics Core Facility at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johannes Fuchs
- Proteomics Core Facility at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Timur Tunovic
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Nina Kamenska
- Norra-Älvsborgs-Länssjukhus (NÄL), Trollhättan, Sweden
| | | | - Yi-Kuo Yu
- National Center for Biotechnology Information (NCBI), Bethesda, MD, United States
| | - Edward R. B. Moore
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Culture Collection University of Gothenburg (CCUG), Sahlgrenska Academy of the University of Gothenburg, Gothenburg, Sweden
| | - Roger Karlsson
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Nanoxis Consulting AB, Gothenburg, Sweden
| |
Collapse
|
13
|
Benamu E, Gajurel K, Anderson JN, Lieb T, Gomez CA, Seng H, Aquino R, Hollemon D, Hong DK, Blauwkamp TA, Kertesz M, Blair L, Bollyky PL, Medeiros BC, Coutre S, Zompi S, Montoya JG, Deresinski S. Plasma Microbial Cell-free DNA Next Generation Sequencing in the Diagnosis and Management of Febrile Neutropenia. Clin Infect Dis 2021; 74:1659-1668. [PMID: 33870413 PMCID: PMC9070798 DOI: 10.1093/cid/ciab324] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Indexed: 11/14/2022] Open
Abstract
Background Standard testing fails to identify a pathogen in most patients with febrile neutropenia (FN). We evaluated the ability of the Karius microbial cell-free DNA sequencing test (KT) to identify infectious etiologies of FN and its impact on antimicrobial management. Methods This prospective study (ClinicalTrials.gov; NCT02912117) enrolled and analyzed 55 patients with FN. Up to 5 blood samples were collected per subject within 24 hours of fever onset (T1) and every 2 to 3 days. KT results were compared with blood culture (BC) and standard microbiological testing (SMT) results. Results Positive agreement was defined as KT identification of ≥1 isolate also detected by BC. At T1, positive and negative agreement were 90% (9/10) and 31% (14/45), respectively; 61% of KT detections were polymicrobial. Clinical adjudication by 3 independent infectious diseases specialists categorized Karius results as: unlikely to cause FN (N = 0); definite (N = 12): KT identified ≥1 organism also found by SMT within 7 days; probable (N = 19): KT result was compatible with a clinical diagnosis; possible (N = 10): KT result was consistent with infection but not considered a common cause of FN. Definite, probable, and possible cases were deemed true positives. Following adjudication, KT sensitivity and specificity were 85% (41/48) and 100% (14/14), respectively. Calculated time to diagnosis was generally shorter with KT (87%). Adjudicators determined real-time KT results could have allowed early optimization of antimicrobials in 47% of patients, by addition of antibacterials (20%) (mostly against anaerobes [12.7%]), antivirals (14.5%), and/or antifungals (3.6%); and antimicrobial narrowing in 27.3% of cases. Clinical Trials Registration NCT02912117 Conclusion KT shows promise in the diagnosis and treatment optimization of FN.
Collapse
Affiliation(s)
- Esther Benamu
- School of Medicine, Division of Infectious Diseases, University of Colorado Denver, Aurora, CO, USA
| | - Kiran Gajurel
- Division of Infectious Diseases, Carolinas Medical Center, Atrium Health, Charlotte, NC, USA
| | - Jill N Anderson
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tullia Lieb
- Hematopoietic Malignancies Unit Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Carlos A Gomez
- Division of Infectious Diseases, Department of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Hon Seng
- Cytovale, Inc, San Francisco, CA, USA
| | | | | | | | | | | | | | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Bruno C Medeiros
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven Coutre
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Jose G Montoya
- The Dr. Jack S. Remington Laboratory for Specialty Diagnostics at the Palo Alto Medical Foundation, Palo Alto, CA, USA
| | - Stan Deresinski
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
14
|
Cheng Q, Parvin B. Rapid identification of a subset of foodborne bacteria in live-cell assays. Appl Microbiol Biotechnol 2020; 104:10571-10584. [PMID: 33185701 DOI: 10.1007/s00253-020-10970-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 09/06/2020] [Accepted: 10/21/2020] [Indexed: 11/28/2022]
Abstract
The detection and identification of microbial pathogens in meat and fresh produce play an essential role in food safety for reducing foodborne illnesses every year. A new approach based on targeting a specific sequence of the 16S rRNA region for each bacterium is proposed and validated. The probe complex consists of a C60, a conjugated RNA detector which targets a specific 16S rRNA sequence, and a complementary fluorescent reporter. The RNA detectors were designed by integrating NIH nucleotide and Vienna RNA Webservice databases, and their specificities were validated by the RDP database. Probe complexes were synthesized for identifying E. coli K12, E. coli O157: H7, S. enterica, Y. enterocolitica, C. perfringens, and L. monocytogenes. First, under controlled conditions of known bacterial mixtures, the efficiency and crosstalk for identifying the foodborne bacteria were quantified to be above 94% and below 5%, respectively. Second, experiments were designed by inoculating meat products by known numbers of bacteria and measuring the limit of detection. In one experiment, 225 g of autoclaved ground chicken was inoculated with 9 E. coli O157:H7, where 6.8 ± 1.2 bacteria with 95% confidence interval were recovered. Third, by positionally printing probe complexes in microwells, specific microorganisms were identified with only one fluorophore. The proposed protocol is a cell-based system, can identify live bacteria in 15 min, requires no amplification, and has the potential to open new surveillance opportunities.Key points• The identification of foodborne bacteria is enabled in live-cell assays.• The limit of detection for 100 g of fresh chicken breast inoculated with 4 bacteria is 2.7 ± 1.4 with 95% confidence interval.• The identification of five bacteria in a coded microwell chip is enabled with only one fluorophore.
Collapse
Affiliation(s)
- Qingsu Cheng
- Department of Electrical and Biomedical Engineering, University of Nevada, Reno, 1664 N. Virginia St., NV, 89557, Reno, USA.,Department of Cell and Molecular Biology, University of Nevada, Reno, 1664 N. Virginia St., Reno, NV, 89557, USA
| | - Bahram Parvin
- Department of Electrical and Biomedical Engineering, University of Nevada, Reno, 1664 N. Virginia St., NV, 89557, Reno, USA. .,Department of Cell and Molecular Biology, University of Nevada, Reno, 1664 N. Virginia St., Reno, NV, 89557, USA.
| |
Collapse
|
15
|
Aistleitner K, Sieper T, Stürz I, Jeske R, Tritscheller S, Mantel S, Tscherne A, Zange S, Stoecker K, Wölfel R. NOTIFy (non-toxic lyophilized field)-FISH for the identification of biological agents by Fluorescence in situ Hybridization. PLoS One 2020; 15:e0230057. [PMID: 32142548 PMCID: PMC7059943 DOI: 10.1371/journal.pone.0230057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/20/2020] [Indexed: 12/02/2022] Open
Abstract
The rapid and reliable diagnostics of highly pathogenic bacteria under restricted field conditions poses one of the major challenges to medical biodefense, especially since false positive or false negative reports might have far-reaching consequences. Fluorescence in situ hybridization (FISH) has the potential to represent a powerful microscopy-based addition to the existing molecular-based diagnostic toolbox. In this study, we developed a set of FISH-probes for the fast, matrix independent and simultaneous detection of thirteen highly pathogenic bacteria in different environmental and clinical sample matrices. Furthermore, we substituted formamide, a routinely used chemical that is toxic and volatile, by non-toxic urea. This will facilitate the application of FISH under resource limited field laboratory conditions. We demonstrate that hybridizations performed with urea show the same specificity and comparable signal intensities for the FISH-probes used in this study. To further simplify the use of FISH in the field, we lyophilized the reagents needed for FISH. The signal intensities obtained with these lyophilized reagents are comparable to freshly prepared reagents even after storage for a month at room temperature. Finally, we show that by the use of non-toxic lyophilized field (NOTIFy)-FISH, specific detection of microorganisms with simple and easily transportable equipment is possible in the field.
Collapse
Affiliation(s)
| | - Tina Sieper
- Bundeswehr Institute of Microbiology, Munich, Germany
| | - Inga Stürz
- Bundeswehr Institute of Microbiology, Munich, Germany
| | - Rimma Jeske
- Bundeswehr Institute of Microbiology, Munich, Germany
| | | | - Sonja Mantel
- Bundeswehr Institute of Microbiology, Munich, Germany
| | | | - Sabine Zange
- Bundeswehr Institute of Microbiology, Munich, Germany
| | - Kilian Stoecker
- Bundeswehr Institute of Microbiology, Munich, Germany
- * E-mail:
| | - Roman Wölfel
- Bundeswehr Institute of Microbiology, Munich, Germany
| |
Collapse
|
16
|
Źródłowski T, Sobońska J, Salamon D, McFarlane IM, Ziętkiewicz M, Gosiewski T. Classical Microbiological Diagnostics of Bacteremia: Are the Negative Results Really Negative? What is the Laboratory Result Telling Us About the "Gold Standard"? Microorganisms 2020; 8:microorganisms8030346. [PMID: 32121353 PMCID: PMC7143506 DOI: 10.3390/microorganisms8030346] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 02/25/2020] [Accepted: 02/27/2020] [Indexed: 12/29/2022] Open
Abstract
Standard blood cultures require at least 24–120 h to be reported as preliminary positive. The objective of this study was to compare the reliability of Gram staining and fluorescent in-situ hybridization (FISH) for detecting bacteria in otherwise negative blood culture bottles. Ninety-six sets were taken from patients with a diagnosis of sepsis. Six incomplete blood culture sets and eight blood cultures sets demonstrating positive growth were excluded. We performed Gram stain and FISH on 82 sets taken from post-operative septic patients: 82 negative aerobic blood cultures, 82 anaerobic blood cultures, and 82 blood samples, as well as 57 blood samples taken from healthy volunteers. From the eighty-two blood sets analyzed from the septic patients, Gram stain visualized bacteria in 62.2% of blood samples, 35.4% of the negative aerobic bottles, and in 31.7% of the negative anaerobic bottles. Utilizing FISH, we detected bacteria in 75.6%, 56.1%, and 64.6% respectively. Among the blood samples from healthy volunteers, FISH detected bacteria in 64.9%, while Gram stain detected bacteria in only 38.6%. The time needed to obtain the study results using Gram stain was 1 h, for FISH 4 h, and for the culture method, considering the duration of growth, 5 days. Gram stain and FISH allow quick detection of bacteria in the blood taken directly from a patient. Finding phagocytosed bacteria, which were also detected among healthy individuals, confirms the hypothesis that blood microbiome exists.
Collapse
Affiliation(s)
- Tomasz Źródłowski
- Thoracic Anesthesia and Respiratory Intensive Care Unit, John Paul II Hospital, 31- 202 Kraków, Poland;
- Department of Internal Medicine, St. John’s Episcopal Hospital, Far Rockaway, NY 11691, USA
| | - Joanna Sobońska
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Krakow, Poland
| | - Dominika Salamon
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Krakow, Poland
| | - Isabel M. McFarlane
- Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Mirosław Ziętkiewicz
- Thoracic Anesthesia and Respiratory Intensive Care Unit, John Paul II Hospital, 31- 202 Kraków, Poland;
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Krakow, Poland
- Correspondence: (M.Z.); (T.G.)
| | - Tomasz Gosiewski
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Krakow, Poland
- Correspondence: (M.Z.); (T.G.)
| |
Collapse
|
17
|
Źródłowski TW, Jurkiewicz-Badacz D, Sroka-Oleksiak A, Salamon D, Bulanda M, Gosiewski T. Comparison of PCR, Fluorescent in Situ Hybridization and Blood Cultures for Detection of Bacteremia in Children and Adolescents During Antibiotic Therapy. Pol J Microbiol 2019; 67:479-486. [PMID: 30550234 PMCID: PMC7256870 DOI: 10.21307/pjm-2018-056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2018] [Indexed: 02/02/2023] Open
Abstract
The gold standard in microbiological diagnostics of bacteremia is a blood culture in automated systems. This method may take several days and has low sensitivity. New screening methods that could quickly reveal the presence of bacteria would be extremely useful. The objective of this study was to estimate the effectiveness of these methods with respect to blood cultures in the context of antibiotic therapy. Blood samples from 92 children with sepsis were analyzed. Blood cultures were carried out in standard automated systems. Subsequently, FISH (Fluorescent In-Situ Hybridization) and nested multiplex-real-time-PCR (PCR) were performed. Blood cultures, FISH and PCR yielded positive results in 18%, 39.1%, and 71.7% of samples, respectively. Significant differences were found between the results obtained through culture before and after induction of antibiotherapy: 25.5% vs. 9.7%. There was no significant difference in FISH and PCR results in relation to antibiotics. The three methods employed demonstrated significant differences in detecting bacteria effectively. Time to obtain test results for FISH and PCR averaged 4–5 hours. FISH and PCR allow to detect bacteria in blood without prior culture. These methods had high sensitivity for the detection of bacteremia regardless of antibiotherapy. They provide more timely results as compared to automated blood culture, and may be useful as rapid screening tests in sepsis.
Collapse
Affiliation(s)
- Tomasz W Źródłowski
- Thoracic Anesthesia and Respiratory Intensive Care Unit, John Paul II Hospital , Cracow , Poland
| | | | - Agnieszka Sroka-Oleksiak
- Chair of Microbiology, Department of Molecular Medical Microbiology, Faculty of Medicine, Jagiellonian University Medical College , Cracow , Poland.,Department of Mycology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College , Czysta 18; 31-121 Krakow , Poland
| | - Dominika Salamon
- Chair of Microbiology, Department of Molecular Medical Microbiology, Faculty of Medicine, Jagiellonian University Medical College , Cracow , Poland
| | - Małgorzata Bulanda
- Chair of Microbiology, Department of Epidemiology of Infection, Faculty of Medicine, Jagiellonian University Medical College , Cracow , Poland
| | - Tomasz Gosiewski
- Chair of Microbiology, Department of Molecular Medical Microbiology, Faculty of Medicine, Jagiellonian University Medical College , Cracow , Poland
| |
Collapse
|
18
|
Oyama N, Winek K, Bäcker-Koduah P, Zhang T, Dames C, Werich M, Kershaw O, Meisel C, Meisel A, Dirnagl U. Exploratory Investigation of Intestinal Function and Bacterial Translocation After Focal Cerebral Ischemia in the Mouse. Front Neurol 2018; 9:937. [PMID: 30510535 PMCID: PMC6254134 DOI: 10.3389/fneur.2018.00937] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/16/2018] [Indexed: 12/13/2022] Open
Abstract
Background and Purpose: The gut communicates with the brain bidirectionally via neural, humoral and immune pathways. All these pathways are affected by acute brain lesions, such as stroke. Brain-gut communication may therefore impact on the overall outcome after CNS-injury. Until now, contradictory reports on intestinal function and translocation of gut bacteria after experimental stroke have been published. Accordingly, we aimed to specifically investigate the effects of transient focal cerebral ischemia on intestinal permeability, gut associated lymphoid tissue and bacterial translocation in an exploratory study using a well-characterized murine stroke model. Methods: After 60 min of middle cerebral artery occlusion (MCAO) we assessed intestinal morphology (time points after surgery day 0, 3, 5, 14, 21) and tight junction protein expression (occludin and claudin-1 at day 1 and 3) in 12-week-old male C57Bl/6J mice. Lactulose/mannitol/sucralose test was performed to assess intestinal permeability 24–72 h after surgery. To investigate the influence of cerebral ischemia on the local immune system of the gut, main immune cell populations in Peyer's patches (PP) were quantified by flow cytometry. Finally, we evaluated bacterial translocation to extraintestinal organs 24 and 72 h after MCAO by microbiological culture and fluorescence in situ hybridization targeting bacterial 16S rRNA. Results: Transient MCAO decreased claudin-1 expression in the ileum but not in the colon. Intestinal morphology (assessed by light microscopy) and permeability did not change measurably after MCAO. After MCAO, animals had significantly fewer B cells in PP compared to naïve mice. Conclusions: In a murine model of stroke, which leads to large brain infarctions in the middle cerebral artery territory, we did not find evidence for overt alterations neither in gut morphology, barrier proteins and permeability nor presence of intestinal bacterial translocation.
Collapse
Affiliation(s)
- Naoki Oyama
- Department of Experimental Neurology, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Katarzyna Winek
- Department of Experimental Neurology, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany.,Neurocure Cluster of Excellence, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Priscilla Bäcker-Koduah
- Department of Experimental Neurology, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany.,Neurocure Cluster of Excellence, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Tian Zhang
- Department of Experimental Neurology, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Claudia Dames
- Institute for Medical Immunology, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Martina Werich
- Medical Department, Division of Hepatology and Gastroenterology, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Olivia Kershaw
- Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Christian Meisel
- Institute for Medical Immunology, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Andreas Meisel
- Department of Experimental Neurology, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany.,Neurocure Cluster of Excellence, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Ulrich Dirnagl
- Department of Experimental Neurology, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany.,Neurocure Cluster of Excellence, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt Universitäts zu Berlin and Berlin Institute of Health, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE), Partner Site Berlin, Berlin, Germany.,QUEST - Center for Transforming Biomedical Research, Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
19
|
Liang F, Browne DJ, Gray MJ, Gartlan KH, Smith DD, Barnard RT, Hill GR, Corrie SR, Markey KA. Development of a Multiplexed Microsphere PCR for Culture-Free Detection and Gram-Typing of Bacteria in Human Blood Samples. ACS Infect Dis 2018; 4:837-844. [PMID: 29350524 DOI: 10.1021/acsinfecdis.7b00277] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bloodstream infection is a significant clinical problem, particularly in vulnerable patient groups such as those undergoing chemotherapy and bone marrow transplantation. Clinical diagnostics for suspected bloodstream infection remain centered around blood culture (highly variable timing, in the order of hours to days to become positive), and empiric use of broad-spectrum antibiotics is therefore employed for patients presenting with febrile neutropenia. Gram-typing provides the first opportunity to target therapy (e.g., combinations containing vancomycin or teicoplanin for Gram-positives; piperacillin-tazobactam or a carbapenem for Gram-negatives); however, current approaches require blood culture. In this study, we describe a multiplexed microsphere-PCR assay with flow cytometry readout, which can distinguish Gram-positive from Gram-negative bacterial DNA in a 3.5 h time period. The combination of a simple assay design (amplicon-dependent release of Gram-type specific Cy3-labeled oligonucleotides) and the Luminex-based readout (for quantifying each specific Cy3-labeled sequence) opens opportunities for further multiplexing. We demonstrate the feasibility of detecting common Gram-positive and Gram-negative organisms after spiking whole bacteria into healthy human blood prior to DNA extraction. Further development of DNA extraction methods is required to reach detection limits comparable to blood culture.
Collapse
Affiliation(s)
- Fang Liang
- Division of Immunology, QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, Queensland 4006, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia campus, Brisbane, Queensland 4072, Australia
| | - Daniel J. Browne
- Division of Immunology, QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, Queensland 4006, Australia
| | - Megan J. Gray
- Division of Immunology, QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, Queensland 4006, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia campus, Brisbane, Queensland 4072, Australia
| | - Kate H. Gartlan
- Division of Immunology, QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, Queensland 4006, Australia
- School of Medicine, The University of Queensland, St Lucia campus, Brisbane, Queensland 4072, Australia
| | - David D. Smith
- Division of Immunology, QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, Queensland 4006, Australia
| | - Ross T. Barnard
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia campus, Brisbane, Queensland 4072, Australia
- Australian Infectious Disease Research Centre, The University of Queensland, St Lucia campus, Brisbane, Queensland 4029, Australia
| | - Geoffrey R. Hill
- Division of Immunology, QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, Queensland 4006, Australia
- Department of Haematology and Bone Marrow Transplantation, The Royal Brisbane and Women’s Hospital, Bowen Bridge Road & Butterfield Street, Brisbane, Queensland 4029, Australia
| | - Simon R. Corrie
- Department of Chemical Engineering, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash and QLD nodes, Monash University, 22 Alliance Lane, Clayton, Victoria 3800, Australia
| | - Kate A. Markey
- Division of Immunology, QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, Queensland 4006, Australia
- School of Medicine, The University of Queensland, St Lucia campus, Brisbane, Queensland 4072, Australia
- Department of Haematology and Bone Marrow Transplantation, The Royal Brisbane and Women’s Hospital, Bowen Bridge Road & Butterfield Street, Brisbane, Queensland 4029, Australia
- Australian Infectious Disease Research Centre, The University of Queensland, St Lucia campus, Brisbane, Queensland 4029, Australia
| |
Collapse
|
20
|
Autophagy Strengthens Intestinal Mucosal Barrier by Attenuating Oxidative Stress in Severe Acute Pancreatitis. Dig Dis Sci 2018; 63:910-919. [PMID: 29427225 DOI: 10.1007/s10620-018-4962-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/01/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Intestinal mucosal barrier dysfunction can be caused by severe acute pancreatitis (SAP). It is normally associated with changes to mucosal autophagy and oxidative stress. OBJECTIVE The aim of this study was to investigate the correlation between autophagy and oxidative stress on the intestinal mucosal barrier of SAP rat model. METHODS SAP was induced by retrograde injection of sodium taurocholate (5%) into the biliopancreatic duct. Bacterial translocation (BT) was detected by 16S rDNA sequencing analysis. Morphological alterations in the pancreas and gut were determined by hematoxylin-eosin staining. Oxidative stress status was determined by measuring the level of intestinal malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione peroxidase (GPx). Western blot, RT-PCR, and immunofluorescent staining were preformed to analyze the expression of tight junction and autophagy proteins. RESULTS According to the sequencing analysis, rats in SAP group were divided into BT (+) group (n = 9) and BT (-) group (n = 8). Pancreatic and intestinal injuries in SAP group were significantly higher than sham operation group. The content of MDA was clearly elevated, and SOD as well as GPx activities were decreased in BT (+) group as compared with BT (-) group. The expression of LC3II and Beclin1 in BT (-) group was higher than that observed in BT (+). In contrast, BT (+) group had a higher level of claudin-2 and a lower level of zonula occluden-1, occludin, and claudin-1. CONCLUSION These results suggest that activated autophagy may attenuate intestinal mucosal barrier dysfunction by preventing and reducing the oxidative stress in SAP.
Collapse
|
21
|
Florio W, Morici P, Ghelardi E, Barnini S, Lupetti A. Recent advances in the microbiological diagnosis of bloodstream infections. Crit Rev Microbiol 2017; 44:351-370. [PMID: 29185372 DOI: 10.1080/1040841x.2017.1407745] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Rapid identification (ID) and antimicrobial susceptibility testing (AST) of the causative agent(s) of bloodstream infections (BSIs) are essential for the prompt administration of an effective antimicrobial therapy, which can result in clinical and financial benefits. Immediately after blood sampling, empirical antimicrobial therapy, chosen on clinical and epidemiological data, is administered. When ID and AST results are available, the clinician decides whether to continue or streamline the antimicrobial therapy, based on the results of the in vitro antimicrobial susceptibility profile of the pathogen. The aim of the present study is to review and discuss the experimental data, advantages, and drawbacks of recently developed technological advances of culture-based and molecular methods for the diagnosis of BSI (including mass spectrometry, magnetic resonance, PCR-based methods, direct inoculation methods, and peptide nucleic acid fluorescence in situ hybridization), the understanding of which could provide new perspectives to improve and fasten the diagnosis and treatment of septic patients. Although blood culture remains the gold standard to diagnose BSIs, newly developed methods can significantly shorten the turnaround time of reliable microbial ID and AST, thus substantially improving the diagnostic yield.
Collapse
Affiliation(s)
- Walter Florio
- a Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia , Università di Pisa , Pisa , Italy
| | - Paola Morici
- a Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia , Università di Pisa , Pisa , Italy
| | - Emilia Ghelardi
- a Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia , Università di Pisa , Pisa , Italy
| | - Simona Barnini
- b U.O. Microbiologia Universitaria Azienda Ospedaliero-Universitaria Pisana , Pisa , Italy
| | - Antonella Lupetti
- a Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia , Università di Pisa , Pisa , Italy
| |
Collapse
|
22
|
Rodolico V, Di Carlo P, Gulotta G, D'Arpa F, Salamone G, Cocorullo G, Agrusa A, Giammanco A, Sergi C. Intra-abdominal Candida spp infection in acute abdomen in a quality assurance (QA)-certified academic setting. J Clin Pathol 2017; 70:579-583. [PMID: 27941028 PMCID: PMC5484093 DOI: 10.1136/jclinpath-2016-203936] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/04/2016] [Accepted: 11/09/2016] [Indexed: 01/24/2023]
Abstract
AIMS To evaluate the contribution of light microscopy to detecting Candida spp infection in patients with complicated intra-abdominal infections (IAIs) admitted for acute abdomen to a quality assurance (QA)-certified surgical emergency ward. METHODS We conducted a retrospective study (2008-2012) of 809 abdominal intraoperative or biopsy tissue specimens obtained from patients admitted with acute abdomen and microbiological samples positive for Candida spp. Demographic data, mortality, comorbidities, specimen type, microscopy results, special histological staining performed, antimicrobial therapy were collected and analysed. Any comments at the multidisciplinary team meeting was recorded in minutes of and approved. RESULTS Sixty-six patients with complicated IAIs due to Candida spp were identified (39 male, 27 female, mean±SD age 75±20 years). Candida albicans was isolated in 35 cases and Candida non-albicans spp in 31 cases. Candida spp were isolated from blood in 50% of all selected microbiological specimens. Patients were stratified according to Candida spp (albicans vs non-albicans), underlying cancer disease and no previous antimicrobial administration, and a positive correlation with C. albicans isolation was found (p=0.009 and p=0.048, respectively). Out of 41 cases with microscopic evaluation, we identified yeast forms, pseudohyphae or both, indicative of Candida spp, in 23. Identification of Candida spp in histological specimens was higher in C. albicans cases than in C. non-albicans cases (73% vs 37.5%). Microscopy allowed prompt treatment of all patients. CONCLUSIONS Light microscopy still has great diagnostic significance, being a solid QA step. It provides rapid information and clues in patients who may harbour impaired defence mechanisms, concurrent chronic conditions and/or cancer.
Collapse
Affiliation(s)
- Vito Rodolico
- Department of Sciences for Health Promotion and Mother & Child Care, Section of Anatomic Pathology, University of Palermo, Palermo, Italy
| | - Paola Di Carlo
- Department of Sciences for Health Promotion and Mother & Child Care, Section of Infectious Disease, University of Palermo, Palermo, Italy
| | - Gaspare Gulotta
- Department of General Surgery and Emergency, University of Palermo, Palermo, Italy
| | - Francesco D'Arpa
- Department of General Surgery and Emergency, University of Palermo, Palermo, Italy
| | - Giuseppe Salamone
- Department of General Surgery and Emergency, University of Palermo, Palermo, Italy
| | - Gianfranco Cocorullo
- Department of General Surgery and Emergency, University of Palermo, Palermo, Italy
| | - Antonino Agrusa
- Department of General Surgery and Emergency, University of Palermo, Palermo, Italy
| | - Anna Giammanco
- Department of Sciences for Health Promotion and Mother & Child Care, Section of Microbiology, University of Palermo, Palermo, Italy
| | - Consolato Sergi
- Department of Lab. Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
- Stollery Children's Hospital, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
23
|
Gosiewski T, Ludwig-Galezowska AH, Huminska K, Sroka-Oleksiak A, Radkowski P, Salamon D, Wojciechowicz J, Kus-Slowinska M, Bulanda M, Wolkow PP. Comprehensive detection and identification of bacterial DNA in the blood of patients with sepsis and healthy volunteers using next-generation sequencing method - the observation of DNAemia. Eur J Clin Microbiol Infect Dis 2016; 36:329-336. [PMID: 27771780 PMCID: PMC5253159 DOI: 10.1007/s10096-016-2805-7] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/26/2016] [Indexed: 12/17/2022]
Abstract
Blood is considered to be a sterile microenvironment, in which bacteria appear only periodically. Previously used methods allowed only for the detection of either viable bacteria with low sensitivity or selected species of bacteria. The Next-Generation Sequencing method (NGS) enables the identification of all bacteria in the sample with their taxonomic classification. We used NGS for the analysis of blood samples from healthy volunteers (n = 23) and patients with sepsis (n = 62) to check whether any bacterial DNA exists in the blood of healthy people and to identify bacterial taxonomic profile in the blood of septic patients. The presence of bacterial DNA was found both in septic and healthy subjects; however, bacterial diversity was significantly different (P = 0.002) between the studied groups. Among healthy volunteers, a significant predominance of anaerobic bacteria (76.2 %), of which most were bacteria of the order Bifidobacteriales (73.0 %), was observed. In sepsis, the majority of detected taxa belonged to aerobic or microaerophilic microorganisms (75.1 %). The most striking difference was seen in the case of Actinobacteria phyla, the abundance of which was decreased in sepsis (P < 0.001) and Proteobacteria phyla which was decreased in the healthy volunteers (P < 0.001). Our research shows that bacterial DNA can be detected in the blood of healthy people and that its taxonomic composition is different from the one seen in septic patients. Detection of bacterial DNA in the blood of healthy people may suggest that bacteria continuously translocate into the blood, but not always cause sepsis; this observation can be called DNAemia.
Collapse
Affiliation(s)
- T Gosiewski
- Department of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| | - A H Ludwig-Galezowska
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, 7C Kopernika Str., 31-034, Krakow, Poland
| | - K Huminska
- Genomic Laboratory, DNA Research Center, Poznan, Poland.,Laboratory of High Throughput Technologies, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - A Sroka-Oleksiak
- Department of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| | - P Radkowski
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, 7C Kopernika Str., 31-034, Krakow, Poland
| | - D Salamon
- Department of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| | | | | | - M Bulanda
- Department of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| | - P P Wolkow
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, 7C Kopernika Str., 31-034, Krakow, Poland.
| |
Collapse
|
24
|
Tziolos N, Giamarellos-Bourboulis EJ. Contemporary approaches to the rapid molecular diagnosis of sepsis. Expert Rev Mol Diagn 2016; 16:1201-1207. [PMID: 27728986 DOI: 10.1080/14737159.2016.1246958] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Although the administration of appropriate antimicrobials within the very first hour remains the mainstay of sepsis management, the correct selection of antimicrobials is hampered by the delay of conventional microbiology providing results after at least 48 hours. Methods of rapid detection of pathogens are an approach to overcome these difficulties. Areas covered: This review analyzes the advantages and the disadvantages of these approaches with major emphasis on technologies based on multiplex PCR for the rapid detection of pathogens using whole blood. The most broadly studied platform is SeptFast. Sensitivity ranges between 42% and 73% and specificity between 50% and 97%. The main disadvantages are high cost, the risk of contamination and the lack of information for the presence of resistance genes. A brief review of the use of PCR techniques for the diagnosis of endocarditis and of the recognition of the bacterial proteome for the rapid identification of grown colonies (MALDI-TOF) is also provided. Expert commentary: More randomized clinical trials are necessary to validate the use of molecular techniques for decision-making for patients' outcomes, taking into consideration the cost-benefit for the patient.
Collapse
Affiliation(s)
- Nikolaos Tziolos
- a 4th Department of Internal Medicine , National and Kapodistrian University of Athens, Medical School , Athens , Greece
| | | |
Collapse
|
25
|
Abstract
Abstract
Background
Critically ill patients are at high risk to suffer from sepsis, even in the absence of an initial infectious source, but the molecular mechanisms for their increased sepsis susceptibility, including a suppressed immune system, remain unclear. Although microbes and pathogen-associated molecular pattern are accepted inducers of sepsis and septic immunosuppression, the role of endogenous Toll-like receptor (TLR) ligands, such as mitochondrial DNA (mtDNA), in altering the immune response is unknown.
Methods
Mitochondrial DNA serum concentrations of the mitochondrial genes D-Loop and adenosine triphosphatase 6 were determined (quantitative polymerase chain reaction) in 165 septic patients and 50 healthy volunteers. Furthermore, cytotoxic T-cell activity was analyzed in wild-type and TLR9 knockout mice, with/without previous mtDNA administration, followed by injection of an ovalbumin-expressing adenoviral vector.
Results
Mitochondrial DNA serum concentrations were increased in septic patients (adenosine triphosphatase 6, 123-fold; D-Loop, 76-fold, P < 0.0001) compared with volunteers. Furthermore, a single mtDNA injection caused profound, TLR9-dependent immunosuppression of adaptive T-cell cytotoxicity in wild-type but not in TLR9 knockout mice and evoked various immunosuppressive mechanisms including the destruction of the splenic microstructure, deletion of cross-presenting dendritic cells, and up-regulation of programmed cell death ligand 1 and indoleamine 2,3-dioxygenase. Several of these findings in mice were mirrored in septic patients, and mtDNA concentrations were associated with an increased 30-day mortality.
Conclusions
The findings of this study imply that mtDNA, an endogenous danger associated molecular pattern, is a hitherto unknown inducer of septic immunoparalysis and one possible link between initial inflammation and subsequent immunosuppression in critically ill patients.
Collapse
|
26
|
Païssé S, Valle C, Servant F, Courtney M, Burcelin R, Amar J, Lelouvier B. Comprehensive description of blood microbiome from healthy donors assessed by 16S targeted metagenomic sequencing. Transfusion 2016; 56:1138-47. [PMID: 26865079 DOI: 10.1111/trf.13477] [Citation(s) in RCA: 298] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/02/2015] [Accepted: 12/03/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Recent studies have revealed that the blood of healthy humans is not as sterile as previously supposed. The objective of this study was to provide a comprehensive description of the microbiome present in different fractions of the blood of healthy individuals. STUDY DESIGN AND METHODS The study was conducted in 30 healthy blood donors to the French national blood collection center (Établissement Français du Sang). We have set up a 16S rDNA quantitative polymerase chain reaction assay as well as a 16S targeted metagenomics sequencing pipeline specifically designed to analyze the blood microbiome, which we have used on whole blood as well as on different blood fractions (buffy coat [BC], red blood cells [RBCs], and plasma). RESULTS Most of the blood bacterial DNA is located in the BC (93.74%), and RBCs contain more bacterial DNA (6.23%) than the plasma (0.03%). The distribution of 16S DNA is different for each fraction and spreads over a relatively broad range among donors. At the phylum level, blood fractions contain bacterial DNA mostly from the Proteobacteria phylum (more than 80%) but also from Actinobacteria, Firmicutes, and Bacteroidetes. At deeper taxonomic levels, there are striking differences between the bacterial profiles of the different blood fractions. CONCLUSION We demonstrate that a diversified microbiome exists in healthy blood. This microbiome has most likely an important physiologic role and could be implicated in certain transfusion-transmitted bacterial infections. In this regard, the amount of 16S bacterial DNA or the microbiome profile could be monitored to improve the safety of the blood supply.
Collapse
Affiliation(s)
| | | | | | | | | | - Jacques Amar
- Vaiomer SAS, Labège.,Department of Therapeutics, Rangueil Hospital, Toulouse, France
| | | |
Collapse
|
27
|
Abstract
Real-time PCR is the traditional face of nucleic acid detection in the diagnostic microbiology laboratory and is now generally regarded as robust enough to be widely adopted. Methods based on nucleic acid detection of this type are bringing increased accuracy to diagnosis in areas where culture is difficult and/or expensive, and these methods are often effective partners to other rapid molecular diagnostic tools such as matrix-assisted laser desorption ionisation-time of flight mass spectrometry (MALDI-TOF MS). This change in practice has particularly affected the recognition of viruses and fastidious or antibiotic-exposed bacteria, but has been also shown to be effective in the recognition of troublesome or specialised phenotypes such as antiviral resistance and transmissible antibiotic resistance in the Enterobacteriaceae. Quantitation and high-intensity sequencing (of multiple whole genomes) has brought new opportunities as well as new challenges to the microbiology community. Diagnostic microbiologists currently training might be expected to deal less with the culture-based techniques of the last half-century than with the high-volume data and complex analyses of the next.
Collapse
|
28
|
Fukumoto H, Sato Y, Hasegawa H, Saeki H, Katano H. Development of a new real-time PCR system for simultaneous detection of bacteria and fungi in pathological samples. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:15479-15488. [PMID: 26823918 PMCID: PMC4713704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/20/2015] [Indexed: 06/05/2023]
Abstract
A novel system for simultaneous detection of pathogenic bacteria and fungi in pathological samples was developed using a real-time polymerase chain reaction (PCR) system. This system, designated the "multi-microbial real-time PCR", has the potential to simultaneously detect 68 bacterial and 9 fungal species in a 96-well plate format. All probe-primer sets were designed to produce amplicons smaller than 210 bp using formalin-fixed paraffin-embedded samples as input. The specificity and sensitivity of each probe-primer set were tested against DNA extracted from pure cultures of specific pathogens. The multi-microbial real-time PCR system revealed profiles of microorganism infection in lung samples collected at autopsy from 10 patients with acquired immunodeficiency syndrome. Staphylococcus aureus was the most common microbe detected (n=8), but with low copy numbers. High copy numbers of Pseudomonas aeruginosa were detected in the lung samples with abscess (n=6). Enterococcus faecium (n=6), Elizabethkingia meningoseptica (n=4), and Candida albicans (n=4) were also frequently detected. In addition, a latent infection of Mycobacterium tuberculosis was detected in one case of pneumonia. In conclusion, this multi-microbial real-time PCR system can be useful for detecting bacteria and fungi in pathological specimens from patients with uncertain diagnoses.
Collapse
Affiliation(s)
- Hitomi Fukumoto
- Department of Pathology, National Institute of Infectious Diseases1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
- Department of Dermatology, Nippon Medical School1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Yuko Sato
- Department of Pathology, National Institute of Infectious Diseases1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Hidehisa Saeki
- Department of Dermatology, Nippon Medical School1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Harutaka Katano
- Department of Pathology, National Institute of Infectious Diseases1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| |
Collapse
|
29
|
Stranieri I, Kanunfre KA, Rodrigues JC, Yamamoto L, Nadaf MIV, Palmeira P, Okay TS. Usefulness of a 16S rDNA real-time PCR to monitor neonatal sepsis and to assist in medical decision to discontinue antibiotics. J Matern Fetal Neonatal Med 2015; 29:2141-4. [PMID: 26334172 PMCID: PMC4841028 DOI: 10.3109/14767058.2015.1077223] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To monitor the bacterial load in newborns with proven infections on the day of admission, 48 h and 7 days after treatment. METHODS Real-time PCR (qPCR) targeting the 16S rDNA. RESULTS The study recruited 17 newborns and the bacterial load was in general low (<50 CFU/mL). In three of four deaths, the bacterial load values increased, and in 11 of the 13 survivors the values decreased until the third evaluation. CONCLUSION Considering the extreme sensitivity and high negative predictive value of qPCR, this test could help to monitor the treatment of neonatal sepsis and to assist in medical decision to discontinue antibiotics.
Collapse
Affiliation(s)
- Inês Stranieri
- a Laboratory of Pathology, Division of Microbiology , Julio Muller University Hospital, Federal University of Mato Grosso , Cuiabá , Brazil
| | - Kelly Aparecida Kanunfre
- b Laboratory of Seroepidemiology and Immunobiology, Institute of Tropical Medicine , University of São Paulo , São Paulo , Brazil .,c Laboratory of Medical Investigation in Immunology (LIM48), Department of Infectious and Parasitic Diseases , School of Medicine, University of São Paulo , São Paulo , Brazil
| | - Jonatas Cristian Rodrigues
- b Laboratory of Seroepidemiology and Immunobiology, Institute of Tropical Medicine , University of São Paulo , São Paulo , Brazil .,c Laboratory of Medical Investigation in Immunology (LIM48), Department of Infectious and Parasitic Diseases , School of Medicine, University of São Paulo , São Paulo , Brazil
| | - Lidia Yamamoto
- b Laboratory of Seroepidemiology and Immunobiology, Institute of Tropical Medicine , University of São Paulo , São Paulo , Brazil
| | | | - Patricia Palmeira
- e Laboratory of Medical Investigation (LIM-36) , Children's Institute, Clinical Hospital, School of Medicine, University of São Paulo , São Paulo , Brazil , and.,f Department of Pediatrics , School of Medicine, University of São Paulo , São Paulo , Brazil
| | - Thelma Suely Okay
- b Laboratory of Seroepidemiology and Immunobiology, Institute of Tropical Medicine , University of São Paulo , São Paulo , Brazil
| |
Collapse
|