1
|
Gleerup D, Trypsteen W, Fraley SI, De Spiegelaere W. Digital PCR in Virology: Current Applications and Future Perspectives. Mol Diagn Ther 2024:10.1007/s40291-024-00751-9. [PMID: 39487879 DOI: 10.1007/s40291-024-00751-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2024] [Indexed: 11/04/2024]
Abstract
Digital PCR (dPCR) has been used in the field of virology since its inception. Technological innovations in microfluidics more than a decade ago caused a sharp increase in its use. There is an emerging consensus that dPCR now outperforms quantitative PCR (qPCR) in the basic parameters such as precision, sensitivity, accuracy, repeatability and resistance to inhibitors. These strengths have led to several current applications in quantification, mutation detection and environmental DNA and RNA samples. In high throughput scenarios, such as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, the cost and throughput still significantly hampered the adaption of dPCR. There is much unexplored potential within the multiplexing capabilities of dPCR. This will allow simultaneous multi-target quantification and can also partially alleviate the throughput and cost drawback. In this review, we discuss the strengths and weaknesses of dPCR with a focus on virology applications and we discuss future applications. Finally, we discuss recent evolutions of the technology in the form of real-time dPCR and digital high-resolution melting.
Collapse
Affiliation(s)
- David Gleerup
- Laboratory of Veterinary Morphology, Faculty of Veterinary Medicine, Ghent University, Campus Merelbeke, Salisburylaan 133, 9820, Merelbeke, Belgium
- Ghent University Digital PCR Consortium, Ghent University, Ghent, Belgium
| | - Wim Trypsteen
- Ghent University Digital PCR Consortium, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, HIV Cure Research Center, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Stephanie I Fraley
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Ward De Spiegelaere
- Laboratory of Veterinary Morphology, Faculty of Veterinary Medicine, Ghent University, Campus Merelbeke, Salisburylaan 133, 9820, Merelbeke, Belgium.
- Ghent University Digital PCR Consortium, Ghent University, Ghent, Belgium.
| |
Collapse
|
2
|
Ajjampur SSR, Mankad S, Manuel M, Ruth R, Prabakaran AD, Janagaraj V, David T, Joseph P, Rupali P. Evaluation of Molecular Assays for Diagnosis of Amoebic Liver Abscess in India with Bayesian Latent Class Analysis. Am J Trop Med Hyg 2024; 111:791-795. [PMID: 39043162 PMCID: PMC11448513 DOI: 10.4269/ajtmh.23-0492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 05/15/2024] [Indexed: 07/25/2024] Open
Abstract
Amoebic liver abscess (ALA) is the most common extra-intestinal complication of Entamoeba histolytica, accounting for 50,000 deaths annually, and is endemic in South Asia. Diagnosis based on microscopic examination is insensitive, and serological assays are not discerning of current infections in endemic settings with high exposure. For a rapid and confirmatory laboratory diagnosis of ALA, the performance of a polymerase chain reaction (PCR), quantitative real time PCR (qPCR), digital droplet PCR (ddPCR), and a loop-mediated isothermal amplification (LAMP) assay that detects E. histolytica DNA in liver abscess pus, and a lectin antigen detection ELISA were evaluated against clinical diagnosis (based on predefined criteria) as the gold standard. Owing to the lack of a laboratory gold standard, a Bayesian latent class analysis approach was also used to determine sensitivity and specificity of these assays. In the latent class analysis, qPCR and ddPCR showed the highest sensitivity (98% and 98.1%) and specificity (both 96.6%), and although clinical diagnosis had a comparable sensitivity to qPCR and ddPCR (95.2%), poorer specificity (64.3%) was seen. Kappa agreement analysis showed that qPCR and ddPCR had a perfect agreement of 1 followed by an agreement of 0.76 (95% CI: 0.64-0.88) with PCR. Considering the performance characteristics and relative ease of setting up qPCR as well as the wide availability of qPCR equipment needed, this would be the most optimal assay for rapid, confirmatory, molecular diagnosis of ALA in the tertiary care laboratory setting in India, whereas further optimization of LAMP or antibody-based detection is required for use at smaller or secondary hospitals.
Collapse
Affiliation(s)
- Sitara Swarna Rao Ajjampur
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Sanket Mankad
- Department of Infectious Diseases, Christian Medical College, Vellore, India
| | - Malathi Manuel
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Renita Ruth
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Ashok D. Prabakaran
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Venkateshprabhu Janagaraj
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Thambu David
- Department of Medicine, Christian Medical College, Vellore, India
| | - Philip Joseph
- Department of Hepatopancreatico Biliary Surgery, Christian Medical College, Vellore, India
| | - Priscilla Rupali
- Department of Infectious Diseases, Christian Medical College, Vellore, India
| |
Collapse
|
3
|
Manuel M, Amato HK, Pilotte N, Chieng B, Araka SB, Siko JEE, Harris M, Nadimpalli ML, Janagaraj V, Houngbegnon P, Rajendiran R, Thamburaj J, Kaliappan SP, Sirois AR, Walch G, Oswald WE, Asbjornsdottir KH, Galagan SR, Walson JL, Williams SA, Luty AJF, Njenga SM, Ibikounlé M, Ajjampur SSR, Pickering AJ. Soil surveillance for monitoring soil-transmitted helminths: Method development and field testing in three countries. PLoS Negl Trop Dis 2024; 18:e0012416. [PMID: 39241051 PMCID: PMC11469484 DOI: 10.1371/journal.pntd.0012416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 10/11/2024] [Accepted: 07/30/2024] [Indexed: 09/08/2024] Open
Abstract
BACKGROUND One-fifth of the global population is infected with soil-transmitted helminths (STH). Mass drug administration (MDA) with deworming medication is widely implemented to control morbidity associated with STH infections. However, surveillance of human infection prevalence by collecting individual stool samples is time-consuming, costly, often stigmatized, and logistically challenging. Current methods of STH detection are poorly sensitive, particularly in low-intensity and low-prevalence populations. METHODOLOGY/PRINCIPAL FINDINGS We aimed to develop a sensitive and specific molecular method for detecting STH DNA in large volumes of soil (20 g) by conducting laboratory and proof of concept studies across field sites in Kenya, Benin, and India. We collected human stool (n = 669) and soil (n = 478) from 322 households across the three study sites. We developed protocols for DNA extraction from 20 g of soil and qPCR to detect Ascaris lumbricoides, Trichuris trichiura, Necator americanus, and Ancylostoma duodenale. Agreement between detection of STH via qPCR, digital droplet PCR (ddPCR), and microscopy-based methods was assessed using the Cohen's Kappa statistic. Finally, we estimated associations between soil characteristics and detection of STH in soil by qPCR, as well as between STH detected in soil and STH detected in stool from matched households, adjusting for soil characteristics. The overall prevalence of STH in soil by qPCR was 31% for A. lumbricoides, 3% for T. trichiura, and 13% for any hookworm species. ddPCR and qPCR performed similarly. However, there was poor agreement between STH detected in soil by qPCR versus light microscopy. Microscopy underestimated the prevalence of A. lumbricoides and N. americanus and overestimated T. trichiura. Detection of an STH species in household soil was strongly associated with increased odds of a household member being infected with that same species. CONCLUSIONS/SIGNIFICANCE Soil surveillance for STH has several benefits over stool-based surveillance, including lower cost and higher success rates for sample collection. Considering that delivery of MDA occurs at the community level, environmental surveillance using molecular methods could be a cost-effective alternate strategy for monitoring STH in these populations.
Collapse
Affiliation(s)
- Malathi Manuel
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College Vellore, Vellore, Tamil Nadu, India
| | - Heather K. Amato
- Department of Civil and Environmental Engineering, University of California, Berkeley, California, United States of America
| | - Nils Pilotte
- Department of Biological Sciences, Quinnipiac University, Hamden, Connecticut, United States of America
| | | | | | - Joël Edoux Eric Siko
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Bénin
- Centre de Recherche pour la lutte contre les Maladies Infectieuses Tropicales (CReMIT/TIDRC), Université d’Abomey-Calavi, Calavi, Bénin
| | - Michael Harris
- Department of Civil and Environmental Engineering, University of California, Berkeley, California, United States of America
| | - Maya L. Nadimpalli
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Venkateshprabhu Janagaraj
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College Vellore, Vellore, Tamil Nadu, India
| | | | - Rajeshkumar Rajendiran
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College Vellore, Vellore, Tamil Nadu, India
| | - Joel Thamburaj
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College Vellore, Vellore, Tamil Nadu, India
| | | | - Allison R. Sirois
- Department of Biological Sciences, Quinnipiac University, Hamden, Connecticut, United States of America
| | - Gretchen Walch
- Smith College, Northampton, Massachusetts, United States of America
| | - William E. Oswald
- Department of Disease Control, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Global Health Division, International Development Group, RTI International, Research Triangle Park, Durham, North Carolina, United States of America
| | - Kristjana H. Asbjornsdottir
- Center of Public Health Sciences, University of Iceland, Reykjavík, Iceland
- DeWorm3, Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Sean R. Galagan
- DeWorm3, Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Judd L. Walson
- DeWorm3, Departments of Global Health, Medicine, Pediatrics and Epidemiology, University of Washington, Seattle, Washington, United States of America
- Departments of International Health, Medicine and Pediatrics, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Steven A. Williams
- Department of Biological Sciences, Smith College, Northampton, Massachusetts, United States of America
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | | | | | - Moudachirou Ibikounlé
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Bénin
- Centre de Recherche pour la lutte contre les Maladies Infectieuses Tropicales (CReMIT/TIDRC), Université d’Abomey-Calavi, Calavi, Bénin
| | - Sitara S. R. Ajjampur
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College Vellore, Vellore, Tamil Nadu, India
| | - Amy J. Pickering
- Department of Civil and Environmental Engineering, University of California, Berkeley, California, United States of America
- Blum Center for Developing Economies, University of California, Berkeley, California, United States of America
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
| |
Collapse
|
4
|
Sancha Dominguez L, Cotos Suárez A, Sánchez Ledesma M, Muñoz Bellido JL. Present and Future Applications of Digital PCR in Infectious Diseases Diagnosis. Diagnostics (Basel) 2024; 14:931. [PMID: 38732345 PMCID: PMC11083499 DOI: 10.3390/diagnostics14090931] [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: 03/19/2024] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
Infectious diseases account for about 3 million deaths per year. The advent of molecular techniques has led to an enormous improvement in their diagnosis, both in terms of sensitivity and specificity and in terms of the speed with which a clinically useful result can be obtained. Digital PCR, or 3rd generation PCR, is based on a series of technical modifications that result in more sensitive techniques, more resistant to the action of inhibitors and capable of direct quantification without the need for standard curves. This review presents the main applications that have been developed for the diagnosis of viral, bacterial, and parasitic infections and the potential prospects for the clinical use of this technology.
Collapse
Affiliation(s)
- Laura Sancha Dominguez
- Department of Microbiology, Hospital Universitario de Salamanca, 37007 Salamanca, Spain; (L.S.D.); (A.C.S.)
- Research Group IIMD-16, Institute for Biomedical Research of Salamanca (IBSAL), SACYL, Universidad de Salamanca, CSIC, 37007 Salamanca, Spain
| | - Ana Cotos Suárez
- Department of Microbiology, Hospital Universitario de Salamanca, 37007 Salamanca, Spain; (L.S.D.); (A.C.S.)
- Research Group IIMD-16, Institute for Biomedical Research of Salamanca (IBSAL), SACYL, Universidad de Salamanca, CSIC, 37007 Salamanca, Spain
| | - María Sánchez Ledesma
- Infectious Diseases Unit, Hospital Universitario de Salamanca, 37007 Salamanca, Spain;
| | - Juan Luis Muñoz Bellido
- Department of Microbiology, Hospital Universitario de Salamanca, 37007 Salamanca, Spain; (L.S.D.); (A.C.S.)
- Research Group IIMD-16, Institute for Biomedical Research of Salamanca (IBSAL), SACYL, Universidad de Salamanca, CSIC, 37007 Salamanca, Spain
- Department of Biomedical and Diagnosis Sciences, Faculty of Medicine, Universidad de Salamanca, 37007 Salamanca, Spain
- Center for Research on Tropical Diseases, Universidad de Salamanca (CIETUS), 37007 Salamanca, Spain
| |
Collapse
|
5
|
Lin Y, Yang J, Wang X, Yang J, Dong L. Establishment of reference measurement procedure and reference material for Treponema pallidum. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:1244-1251. [PMID: 38319302 DOI: 10.1039/d3ay01906c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Stem cell preparations, as a new type of biotherapeutic product, should be subject to strict quality control in terms of cell safety. The testing of stem cell donors and blood products used in stem cell cultures, including but not limited to Treponema pallidum, is needed to reduce the risk of transmission of infectious diseases by stem cell medical products. In this study, a reference measurement procedure (RMP) was established based on digital PCR (dPCR). A homogeneous reference material (RM) of TP containing the tpp47 gene has been developed and characterized. Two dPCR assays (A and B) show ideal linearity within five orders of magnitude. The limit of quantification (LoQ) for both assays is 57 copies/reaction; the limits of detection (LoD) are 9.69 and 9.59 copies/reaction, respectively. The quantitative results of the established duplex dPCR assay are in good agreement. The RM of TP containing the tpp47 gene has been developed and characterized. The reference value with its expanded uncertainty is (2.21 ± 0.22) × 106 copies per μL determined by the established dPCR RMP. The developed dPCR was validated by applying a simulated stem cell matrix, and no impact was observed on the accuracy of dPCR. By providing an accurate reference value for the absolute copy number of the target gene, the developed RM can be used to improve the reliability of TP testing in the production of stem cell preparations and clinical diagnostics.
Collapse
Affiliation(s)
- Yanmin Lin
- College of Food Science and Technology, Shanghai Ocean University, 201306, Shanghai, P. R. China.
- Center for Advanced Measurement of Science, National Institute of Metrology, 100029, Beijing, P. R. China.
| | - Jiayi Yang
- Center for Advanced Measurement of Science, National Institute of Metrology, 100029, Beijing, P. R. China.
| | - Xia Wang
- Center for Advanced Measurement of Science, National Institute of Metrology, 100029, Beijing, P. R. China.
| | - Jingya Yang
- College of Food Science and Technology, Shanghai Ocean University, 201306, Shanghai, P. R. China.
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, 201306, Shanghai, P. R. China
| | - Lianhua Dong
- Center for Advanced Measurement of Science, National Institute of Metrology, 100029, Beijing, P. R. China.
| |
Collapse
|
6
|
Porco D, Purnomo CA, Glesener L, Proess R, Lippert S, Jans K, Colling G, Schneider S, Stassen R, Frantz AC. eDNA-based monitoring of Batrachochytrium dendrobatidis and Batrachochytrium salamandrivorans with ddPCR in Luxembourg ponds: taking signals below the Limit of Detection (LOD) into account. BMC Ecol Evol 2024; 24:4. [PMID: 38178008 PMCID: PMC10768104 DOI: 10.1186/s12862-023-02189-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/09/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Batrachochytrium dendrobatidis (Bd) and Batrachochytrium salamandrivorans (Bsal) are two pathogenic fungi that are a significant threat to amphibian communities worldwide. European populations are strongly impacted and the monitoring of the presence and spread of these pathogens is crucial for efficient decision-making in conservation management. RESULTS Here we proposed an environmental DNA (eDNA) monitoring of these two pathogenic agents through droplet digital PCR (ddPCR) based on water samples from 24 ponds in Luxembourg. In addition, amphibians were swabbed in eight of the targeted ponds in order to compare the two approaches at site-level detection. This study allowed the development of a new method taking below-Limit of Detection (LOD) results into account thanks to the statistical comparison of the frequencies of false positives in no template controls (NTC) and below-LOD results in technical replicates. In the eDNA-based approach, the use of this method led to an increase in Bd and Bsal detection of 28 and 50% respectively. In swabbing, this resulted in 8% more positive results for Bd. In some samples, the use of technical replicates allowed to recover above-LOD signals and increase Bd detection by 35 and 33% respectively for eDNA and swabbing, and Bsal detection by 25% for eDNA. CONCLUSIONS These results confirmed the usefulness of technical replicates to overcome high levels of stochasticity in very low concentration samples even for a highly sensitive technique such as ddPCR. In addition, it showed that below-LOD signals could be consistently recovered and the corresponding amplification events assigned either to positive or negative detection via the method developed here. This methodology might be particularly worth pursuing in pathogenic agents' detection as false negatives could have important adverse consequences. In total, 15 ponds were found positive for Bd and four for Bsal. This study reports the first record of Bsal in Luxembourg.
Collapse
Affiliation(s)
- David Porco
- Musée national d'histoire naturelle du Luxembourg, 25, rue Münster, Luxembourg, L-2160, Luxembourg.
- Fondation Faune Flore, 24, rue Münster, Luxembourg, L-2160, Luxembourg.
| | - Chanistya Ayu Purnomo
- Musée national d'histoire naturelle du Luxembourg, 25, rue Münster, Luxembourg, L-2160, Luxembourg
| | - Liza Glesener
- Naturschutzsyndikat SICONA, 12, rue de Capellen, L-8393 Olm, Luxembourg, Luxembourg
| | - Roland Proess
- Umweltplanungsbüro Ecotop, 45, Schlassuecht, L-7435 Hollenfels, Luxembourg, Luxembourg
| | - Stéphanie Lippert
- Musée national d'histoire naturelle du Luxembourg, 25, rue Münster, Luxembourg, L-2160, Luxembourg
| | - Kevin Jans
- Natur&ëmwelt Fondation Hëllef fir d'Natur, 5, Route de Luxembourg, L-1899, Kockelscheuer, Luxembourg
| | - Guy Colling
- Musée national d'histoire naturelle du Luxembourg, 25, rue Münster, Luxembourg, L-2160, Luxembourg
- Fondation Faune Flore, 24, rue Münster, Luxembourg, L-2160, Luxembourg
| | - Simone Schneider
- Musée national d'histoire naturelle du Luxembourg, 25, rue Münster, Luxembourg, L-2160, Luxembourg
- Naturschutzsyndikat SICONA, 12, rue de Capellen, L-8393 Olm, Luxembourg, Luxembourg
| | - Raf Stassen
- Biota.lu, 9a, Rue Principale, L-6990, Hostert, Luxembourg
| | - Alain C Frantz
- Musée national d'histoire naturelle du Luxembourg, 25, rue Münster, Luxembourg, L-2160, Luxembourg
- Fondation Faune Flore, 24, rue Münster, Luxembourg, L-2160, Luxembourg
| |
Collapse
|
7
|
Reynolds DE, Pan M, Yang J, Galanis G, Roh YH, Morales RT, Kumar SS, Heo S, Xu X, Guo W, Ko J. Double Digital Assay for Single Extracellular Vesicle and Single Molecule Detection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303619. [PMID: 37802976 PMCID: PMC10667851 DOI: 10.1002/advs.202303619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 09/13/2023] [Indexed: 10/08/2023]
Abstract
Extracellular vesicles (EVs) have emerged as a promising source of biomarkers for disease diagnosis. However, current diagnostic methods for EVs present formidable challenges, given the low expression levels of biomarkers carried by EV samples, as well as their complex physical and biological properties. Herein, a highly sensitive double digital assay is developed that allows for the absolute quantification of individual molecules from a single EV. Because the relative abundance of proteins is low for a single EV, tyramide signal amplification (TSA) is integrated to increase the fluorescent signal readout for evaluation. With the integrative microfluidic technology, the technology's ability to compartmentalize single EVs is successfully demonstrated, proving the technology's digital partitioning capacity. Then the device is applied to detect single PD-L1 proteins from single EVs derived from a melanoma cell line and it is discovered that there are ≈2.7 molecules expressed per EV, demonstrating the applicability of the system for profiling important prognostic and diagnostic cancer biomarkers for therapy response, metastatic status, and tumor progression. The ability to accurately quantify protein molecules of rare abundance from individual EVs will shed light on the understanding of EV heterogeneity and discovery of EV subtypes as new biomarkers.
Collapse
Affiliation(s)
- David E. Reynolds
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Menghan Pan
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Jingbo Yang
- Department of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - George Galanis
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Yoon Ho Roh
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | | | | | - Su‐Jin Heo
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Department of Orthopaedic SurgeryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Xiaowei Xu
- Department of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Wei Guo
- Department of BiologySchool of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Jina Ko
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Department of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| |
Collapse
|
8
|
Shahi N, Prasartset T, Surachetpong W. A specific and sensitive droplet digital polymerase chain reaction assay for the detection of tilapia lake virus in fish tissue and environmental samples. JOURNAL OF FISH DISEASES 2023; 46:957-966. [PMID: 37294665 DOI: 10.1111/jfd.13816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/11/2023]
Abstract
Tilapia lake virus (TiLV) causes high mortality in farmed and wild tilapia in various countries. We developed a highly specific and sensitive droplet digital polymerase chain reaction (ddPCR) assay to detect and quantify TiLV. The ddPCR assay could detect the virus at a lower threshold than the reverse transcription-quantitative polymerase reaction (RT-qPCR) method, and the sensitivity of the ddPCR assay was 10-fold higher. The diagnostic sensitivity and specificity of the ddPCR assay were 100% and did not cross-react with tilapia tissues infected with Tilapia parvovirus, Infectious spleen and kidney necrosis virus, Aeromonas hydrophila, Streptococcus agalactiae, S. iniae and Francisella noatunensis. The assay reproducibility was demonstrated by a high correlation coefficient of 0.998, and the inter-assay coefficients of variability indicated that the ddPCR assay exhibited low variability within and between measurements. The detection limit of the TiLV ddPCR assay was 100 fg cDNA, which is equal to 3.3 copies of TiLV. Furthermore, the ddPCR assay could detect TiLV in mucus, water and infected tissue samples and the lowest copy number of TiLV detected in water samples by the ddPCR assay was 7.9 ± 0.99 copies/reaction The results of the clinical samples tested for TiLV revealed that the ddPCR assay had a relatively higher detection rate than the RT-qPCR method. Overall, the ddPCR method offers a highly promising approach for the absolute quantification of TiLV in carrier fish and samples from the environment with low viral concentrations.
Collapse
Affiliation(s)
- Neetu Shahi
- Department of Veterinary Microbiology and Immunology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Tharinthon Prasartset
- Department of Veterinary Microbiology and Immunology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Win Surachetpong
- Department of Veterinary Microbiology and Immunology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| |
Collapse
|
9
|
Guterres A. Viral load: We need a new look at an old problem? J Med Virol 2023; 95:e29061. [PMID: 37638475 DOI: 10.1002/jmv.29061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/22/2023] [Accepted: 08/17/2023] [Indexed: 08/29/2023]
Abstract
The concept of viral load was introduced in the 1980s to measure the amount of viral genetic material in a person's blood, primarily for human immunodeficiency virus (HIV). It has since become crucial for monitoring HIV infection progression and assessing the efficacy of antiretroviral therapy. However, during the coronavirus disease 2019 pandemic, the term "viral load" became widely popularized, not only for the scientific community but for the general population. Viral load plays a critical role in both clinical patient management and research, providing valuable insights for antiviral treatment strategies, vaccination efforts, and epidemiological control measures. As measuring viral load is so important, why don't researchers discuss the best way to do it? Is it simply acceptable to use raw Ct values? Relying solely on Ct values for viral load estimation can be problematic due to several reasons. First, Ct values can vary between different quantitative polymerase chain reaction assays, platforms, and laboratories, making it difficult to compare data across studies. Second, Ct values do not directly measure the quantity of viral particles in a sample and they can be influenced by various factors such as initial viral load, sample quality, and assay sensitivity. Moreover, variations in viral RNA extraction and reverse-transcription steps can further impact the accuracy of viral load estimation, emphasizing the need for careful interpretation of Ct values in viral load assessment. Interestingly, we did not observe scientific articles addressing different strategies to quantify viral load. The absence of standardized and validated methods impedes the implementation of viral load monitoring in clinical management. The variability in cell quantities within samples and the variation in viral particle numbers within infected cells further challenge accurate viral load measurement and interpretation. To advance the field and improve patient outcomes, there is an urgent need for the development and validation of tailored, standardized methods for precise viral load quantification.
Collapse
Affiliation(s)
- Alexandro Guterres
- Laboratório de Hantaviroses e Rickettsioses, Instituto Oswaldo Cruz Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos, Vice-Diretoria de Desenvolvimento Tecnológico, Bio-Manguinhos, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| |
Collapse
|
10
|
Cao G, Xiong Y, Shi M, Qiu Y, Bian Y, Nie F, Huo D, Hou C. The End of the Gray Zone: One-Tube Nested Recombinase Polymerase Amplification with Ultrahigh Signal-to-Noise Ratio for Precisely Detecting and Surveilling Viruses. Anal Chem 2023. [PMID: 37367936 DOI: 10.1021/acs.analchem.3c01609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
The samples were difficult to accurately determine positive or negative between 35 and 40 cycles by real-time quantitative PCR (qPCR) as the standard method. Here, we developed one-tube nested recombinase polymerase amplification (ONRPA) technology with CRISPR/Cas12a to overcome this difficulty. ONRPA broke the amplification plateau to substantially enhance the signals, which considerably improved the sensitivity and eliminated the problem of gray area. Using two pairs of primers one after another, it improved precision by lowering the probability of magnifying several target zones, which was completely free of contamination by nonspecific amplification. This was important in nucleic acid testing. Finally, by the CRISPR/Cas12a system as a terminal output, the approach achieved a high signal output as few as 2.169 copies·μL-1 in 32 min. ONRPA was 100-fold more sensitive than conventional RPA and 1000-fold compared to qPCR. ONRPA coupled with CRISPR/Cas12a will be an important and new promoter of RPA in clinical applications.
Collapse
Affiliation(s)
- Gaihua Cao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, Sichuan 400044, PR China
- State Key Laboratory of Cattle Diseases Detection (Chongqing) of Customs. Diagnosis and Testing Laboratory of Lumpy Skin Disease, Chongqing Customs Technology Center, Chongqing 400020, PR China
| | - Yifan Xiong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, Sichuan 400044, PR China
- State Key Laboratory of Cattle Diseases Detection (Chongqing) of Customs. Diagnosis and Testing Laboratory of Lumpy Skin Disease, Chongqing Customs Technology Center, Chongqing 400020, PR China
| | - Meimei Shi
- State Key Laboratory of Cattle Diseases Detection (Chongqing) of Customs. Diagnosis and Testing Laboratory of Lumpy Skin Disease, Chongqing Customs Technology Center, Chongqing 400020, PR China
| | - Yue Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, Sichuan 400044, PR China
- State Key Laboratory of Cattle Diseases Detection (Chongqing) of Customs. Diagnosis and Testing Laboratory of Lumpy Skin Disease, Chongqing Customs Technology Center, Chongqing 400020, PR China
| | - Yong Bian
- Science and Technology Research Center of China Customs, Beijing 100730, PR China
| | - Fuping Nie
- State Key Laboratory of Cattle Diseases Detection (Chongqing) of Customs. Diagnosis and Testing Laboratory of Lumpy Skin Disease, Chongqing Customs Technology Center, Chongqing 400020, PR China
| | - Danqun Huo
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, Sichuan 400044, PR China
| | - Changjun Hou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, Sichuan 400044, PR China
- Chongqing Key Laboratory of Bio-perception & Intelligent Information Processing, School of Microelectronics and Communication Engineering, Chongqing University, Chongqing, Sichuan 400044, PR China
| |
Collapse
|
11
|
Zerbato JM, Avihingsanon A, Singh KP, Zhao W, Deleage C, Rosen E, Cottrell ML, Rhodes A, Dantanarayana A, Tumpach C, Tennakoon S, Crane M, Price DJ, Braat S, Mason H, Roche M, Kashuba AD, Revill PA, Audsley J, Lewin SR. HIV DNA persists in hepatocytes in people with HIV-hepatitis B co-infection on antiretroviral therapy. EBioMedicine 2022; 87:104391. [PMID: 36502576 PMCID: PMC9763386 DOI: 10.1016/j.ebiom.2022.104391] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/04/2022] [Accepted: 11/17/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND HIV can infect multiple cells in the liver including hepatocytes, Kupffer cells and infiltrating T cells, but whether HIV can persist in the liver in people with HIV (PWH) on suppressive antiretroviral therapy (ART) remains unknown. METHODS In a prospective longitudinal cohort of PWH and hepatitis B virus (HBV) co-infection living in Bangkok, Thailand, we collected blood and liver biopsies from 18 participants prior to and following ART and quantified HIV and HBV persistence using quantitative (q)PCR and RNA/DNAscope. Antiretroviral (ARV) drug levels were quantified using mass spectroscopy. FINDINGS In liver biopsies taken prior to ART, HIV DNA and HIV RNA were detected by qPCR in 53% (9/17) and 47% (8/17) of participants respectively. Following a median ART duration of 3.4 years, HIV DNA was detected in liver in 61% (11/18) of participants by either qPCR, DNAscope or both, but only at very low and non-quantifiable levels. Using immunohistochemistry, HIV DNA was observed in both hepatocytes and liver infiltrating CD4+ T cells on ART. HIV RNA was not detected in liver biopsies collected on ART, by either qPCR or RNAscope. All ARVs were clearly detected in liver tissue. INTERPRETATION Persistence of HIV DNA in liver in PWH on ART represents an additional reservoir that warrants further investigation. FUNDING National Health and Medical Research Council of Australia (Project Grant APP1101836, 1149990, and 1135851); This project has been funded in part with federal funds from the National Cancer Institute, National Institutes of Health, under Contract No. 75N91019D00024.
Collapse
Affiliation(s)
- Jennifer M. Zerbato
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Anchalee Avihingsanon
- HIV-NAT, Thai Red Cross AIDS Research Centre and Centre of Excellence in Tuberculosis, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kasha P. Singh
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia,Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia,Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | - Wei Zhao
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Claire Deleage
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Elias Rosen
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | | | - Ajantha Rhodes
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Ashanti Dantanarayana
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Carolin Tumpach
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Surekha Tennakoon
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Megan Crane
- National Centre for Infections in Cancer, Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - David J. Price
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia,Centre for Epidemiology & Biostatistics, Melbourne School of Population & Global Health, University of Melbourne, Melbourne, Australia
| | - Sabine Braat
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia,Centre for Epidemiology & Biostatistics, Melbourne School of Population & Global Health, University of Melbourne, Melbourne, Australia,MISCH (Methods and Implementation Support for Clinical Health) Research Hub, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Hugh Mason
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at The Peter Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Michael Roche
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Angela D.M. Kashuba
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Peter A. Revill
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at The Peter Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Jennifer Audsley
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Sharon R. Lewin
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia,Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia,Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia,Corresponding author. Department of Infectious Diseases, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, 786-798 Elizabeth Street, Melbourne, Victoria 3010, Australia.
| |
Collapse
|
12
|
Porco D, Hermant S, Purnomo CA, Horn M, Marson G, Colling G. Getting rid of ‘rain’ and ‘stars’: Mitigating inhibition effects on ddPCR data analysis, the case study of the invasive crayfish Pacifastacus leniusculus in the streams of Luxembourg. PLoS One 2022; 17:e0275363. [PMCID: PMC9668142 DOI: 10.1371/journal.pone.0275363] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
ddPCR is becoming one of the most widely used tool in the field of eDNA-based aquatic monitoring. Although emulsion PCR used in ddPCR confers a partial mitigation to inhibition due to the high number of reactions for a single sample (between 10K and 20K), it is not impervious to it. Our results showed that inhibition impacts the amplitude of fluorescence in positive droplets with a different intensity among rivers. This signal fluctuation could jeopardize the use of a shared threshold among samples from different origin, and thus the accurate assignment of the positive droplets which is particularly important for low concentration samples such as eDNA ones: amplification events are scarce, thus their objective discrimination as positive is crucial. Another issue, related to target low concentration, is the artifactual generation of high fluorescence droplets (‘stars’). Indeed, these could be counted as positive with a single threshold solution, which in turn could produce false positive and incorrect target concentration assessments. Approximating the positive and negative droplets distribution as normal, we proposed here a double threshold method accounting for both high fluorescence droplets (‘stars’) and PCR inhibition impact in delineating positive droplets clouds. In the context of low concentration template recovered from environmental samples, the application of this method of double threshold establishment could allow for a consistent sorting of the positive and negative droplets throughout ddPCR data generated from samples with varying levels of inhibitor contents. Due to low concentrations template and inhibition effects, Quantasoft software produced an important number of false negatives and positive comparatively to the double threshold method developed here. This case study allowed the detection of the invasive crayfish P. leniusculus in 32 out of 34 sampled sites from two main rivers (Alzette and Sûre) and five of their tributaries (Eisch, Attert, Mamer, Wiltz and Clerve).
Collapse
Affiliation(s)
- David Porco
- Musée National d’histoire Naturelle, Life Science Department—Invertebrate Zoology, Population Biology and Evolution, Luxembourg, Luxembourg
- Fondation Faune Flore, Luxembourg, Luxembourg
- * E-mail:
| | - Sylvie Hermant
- Musée National d’histoire Naturelle, Life Science Department—Invertebrate Zoology, Population Biology and Evolution, Luxembourg, Luxembourg
| | - Chanistya Ayu Purnomo
- Musée National d’histoire Naturelle, Life Science Department—Invertebrate Zoology, Population Biology and Evolution, Luxembourg, Luxembourg
- Fondation Faune Flore, Luxembourg, Luxembourg
| | - Mario Horn
- Musée National d’histoire Naturelle, Life Science Department—Invertebrate Zoology, Population Biology and Evolution, Luxembourg, Luxembourg
| | - Guy Marson
- Musée National d’histoire Naturelle, Life Science Department—Invertebrate Zoology, Population Biology and Evolution, Luxembourg, Luxembourg
| | - Guy Colling
- Musée National d’histoire Naturelle, Life Science Department—Invertebrate Zoology, Population Biology and Evolution, Luxembourg, Luxembourg
| |
Collapse
|
13
|
Le Duff Y, Gärtner K, Busby EJ, Dalzini A, Danaviah S, Fuentes JLJ, Giaquinto C, Huggett JF, Hurley M, Marcellin AG, Muñoz-Fernández MÁ, O’Sullivan DM, Persaud D, Powell L, Rigsby P, Rossi P, de Rossi A, Siems L, Smit T, Watters SA, Almond N, Nastouli E. Assessing the Variability of Cell-Associated HIV DNA Quantification through a Multicenter Collaborative Study. Microbiol Spectr 2022; 10:e0024322. [PMID: 35658711 PMCID: PMC9241949 DOI: 10.1128/spectrum.00243-22] [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: 02/03/2022] [Accepted: 04/20/2022] [Indexed: 11/20/2022] Open
Abstract
Reliable and accurate quantification of cell-associated HIV DNA (CA HIV DNA) is critical for early infant diagnosis, clinical management of patients under therapy, and to inform new therapeutics efficacy. The present study assessed the variability of CA HIV DNA quantification obtained from various assays and the value of using reference materials to help harmonize the measurements. Using a common set of reagents, our multicenter collaborative study highlights significant variability of CA HIV DNA quantification and lower limit of quantification across assays. The quantification of CA HIV DNA from a panel of infected PBMCs can be harmonized through cross-subtype normalization but assay calibration with the commonly used 8E5 cell line failed to reduce quantification variability between assays, demonstrating the requirement to thoroughly evaluate reference material candidates to help improve the comparability of CA HIV DNA diagnostic assay performance. IMPORTANCE Despite a global effort, HIV remains a major public health burden with an estimated 1.5 million new infections occurring in 2020. HIV DNA is an important viral marker, and its monitoring plays a critical role in the fight against HIV: supporting diagnosis in infants and underpinning clinical management of patients under therapy. Our study demonstrates that HIV DNA measurement of the same samples can vary significantly from one laboratory to another, due to heterogeneity in the assay, protocol, and reagents used. We show that when carefully selected, reference materials can reduce measurement variability and harmonize HIV DNA quantification across laboratories, which will help contribute to improved diagnosis and clinical management of patients living with HIV.
Collapse
Affiliation(s)
- Yann Le Duff
- Division of Infectious Disease Diagnostics, Centre for AIDS Reagent, National Institute for Biological Standards and Control, South Mimms, United Kingdom
| | - Kathleen Gärtner
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Eloise J. Busby
- National Measurement Laboratory, LGC group Teddington, Middlesex, United Kingdom
| | - Annalisa Dalzini
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | | | - José Luis Jiménez Fuentes
- Instituto Investigación Sanitaria Gregorio Marañón, Laboratorio InmunoBiología Molecular and Spanish HIV HGM BioBank, Madrid, Spain
| | - Carlo Giaquinto
- Department for Woman’s and Child’s Health, University of Padova, Padua, Italy
| | - Jim F. Huggett
- National Measurement Laboratory, LGC group Teddington, Middlesex, United Kingdom
| | - Matthew Hurley
- Division of Infectious Disease Diagnostics, Centre for AIDS Reagent, National Institute for Biological Standards and Control, South Mimms, United Kingdom
| | - Anne-Geneviève Marcellin
- Sorbonne Université, INSERM, Institut Pierre Louis d’Epidémiologie et de Santé Publique (IPLESP), Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié Salpêtrière Hospital, Department of Virology, Paris, France
| | - María Ángeles Muñoz-Fernández
- Instituto Investigación Sanitaria Gregorio Marañón, Laboratorio InmunoBiología Molecular and Spanish HIV HGM BioBank, Madrid, Spain
| | - Denise M. O’Sullivan
- National Measurement Laboratory, LGC group Teddington, Middlesex, United Kingdom
| | - Deborah Persaud
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Laura Powell
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peter Rigsby
- Division of Analytical Biological Sciences, National Institute for Biological Standards and Control, South Mimms, United Kingdom
| | - Paolo Rossi
- Department of Pediatrics, University of Rome Tor Vergata, Rome, Italy
| | - Anita de Rossi
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Lilly Siems
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Theresa Smit
- Africa Health Research Institute, Durban, South Africa
| | - Sarah A. Watters
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Neil Almond
- Division of Infectious Disease Diagnostics, Centre for AIDS Reagent, National Institute for Biological Standards and Control, South Mimms, United Kingdom
| | - Eleni Nastouli
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
14
|
Zhang Y, Zhao Y, Cole T, Zheng J, Bayinqiaoge, Guo J, Tang SY. Microfluidic flow cytometry for blood-based biomarker analysis. Analyst 2022; 147:2895-2917. [PMID: 35611964 DOI: 10.1039/d2an00283c] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Flow cytometry has proven its capability for rapid and quantitative analysis of individual cells and the separation of targeted biological samples from others. The emerging microfluidics technology makes it possible to develop portable microfluidic diagnostic devices for point-of-care testing (POCT) applications. Microfluidic flow cytometry (MFCM), where flow cytometry and microfluidics are combined to achieve similar or even superior functionalities on microfluidic chips, provides a powerful single-cell characterisation and sorting tool for various biological samples. In recent years, researchers have made great progress in the development of the MFCM including focusing, detecting, and sorting subsystems, and its unique capabilities have been demonstrated in various biological applications. Moreover, liquid biopsy using blood can provide various physiological and pathological information. Thus, biomarkers from blood are regarded as meaningful circulating transporters of signal molecules or particles and have great potential to be used as non (or minimally)-invasive diagnostic tools. In this review, we summarise the recent progress of the key subsystems for MFCM and its achievements in blood-based biomarker analysis. Finally, foresight is offered to highlight the research challenges faced by MFCM in expanding into blood-based POCT applications, potentially yielding commercialisation opportunities.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Ying Zhao
- National Chengdu Centre of Safety Evaluation of Drugs, West China Hospital of Sichuan University, Chengdu, China
| | - Tim Cole
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Jiahao Zheng
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Bayinqiaoge
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Jinhong Guo
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Shi-Yang Tang
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
15
|
Renault C, Bolloré K, Pisoni A, Motto-Ros C, Van de Perre P, Reynes J, Tuaillon E. Accuracy of real-time PCR and digital PCR for the monitoring of total HIV DNA under prolonged antiretroviral therapy. Sci Rep 2022; 12:9323. [PMID: 35665775 PMCID: PMC9167282 DOI: 10.1038/s41598-022-13581-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 04/21/2022] [Indexed: 12/16/2022] Open
Abstract
Total HIV DNA is a standard marker to monitor the HIV reservoir in people living with HIV. We investigated HIV DNA quantification accuracy by a real-time PCR kit (qPCR) and digital PCR (dPCR) method within the same set of primers and probes. Among 48 aviremic patients followed for up to 7 years with qPCR, the mean coefficient of variation of total HIV DNA between two successive measurements was 77% (± 0.42log10 HIVDNA copies/106 PBMC). The total HIV DNA quantified by the two PCR methods has a high correlation (0.99 and 0.83, for 8E5 and PLHIV samples, respectively), but we observed better repeatability and reproducibility of the dPCR compared to the qPCR (CV of 11.9% vs. 24.7% for qPCR, p-value = 0.024). Furthermore, we highlighted a decay of the number of HIV copies in the 8E5 cell line qPCR standard over time (from 0.73 to 0.43 copies per cell), contributing to variations of HIV DNA results in patients whose HIV reservoir should be theoretically stabilized. Our study highlighted that absolute quantification of total HIV DNA by dPCR allows more accurate monitoring of the HIV reservoir than qPCR in patients under prolonged antiretroviral therapy.
Collapse
Affiliation(s)
- Constance Renault
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang, Antilles University, Montpellier, France
| | - Karine Bolloré
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang, Antilles University, Montpellier, France
| | - Amandine Pisoni
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang, Antilles University, Montpellier, France.,CHU de Montpellier, Montpellier, France
| | - Camille Motto-Ros
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang, Antilles University, Montpellier, France
| | - Philippe Van de Perre
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang, Antilles University, Montpellier, France.,CHU de Montpellier, Montpellier, France
| | - Jacques Reynes
- IRD UMI 233, INSERM U1175, Montpellier University, Montpellier, France.,Infectious Diseases Department, CHU de Montpellier, Montpellier, France
| | - Edouard Tuaillon
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang, Antilles University, Montpellier, France. .,CHU de Montpellier, Montpellier, France.
| |
Collapse
|
16
|
Wang HJ, Xiang YH, Hu R, Ji R, Wang YP. Research progress in laboratory detection of SARS-CoV-2. Ir J Med Sci 2022; 191:509-517. [PMID: 33763777 PMCID: PMC7990494 DOI: 10.1007/s11845-021-02604-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/12/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Nucleic acid testing is a reliable method for diagnosing viral infection in clinical samples. However, when the number of cases is huge and there are individual differences in the virus itself, the probability of false-negative results increases. With the advancement in research on the new coronavirus, new detection technologies that use serum-specific antibodies as detection targets have been developed. These detection technologies have high efficiency and shorter turnaround time, which ultimately shortens the time required for diagnosis. This article summarizes the methods that have been reported to date for the detection of the new coronavirus and discusses their principles and technical characteristics. AIMS Compare the advantages and disadvantages of various SARS-CoV-2 detection methods and analyze their principles. METHODS Searched reports on SARS-CoV-2 detection methods published so far, extracted the data and analyzed them. Use the primer blast function of NCBI to analyze the primers used in qRT-PCR detection. RESULTS The detection sensitivity was the highest when nucleocapsid protein gene was used as the target, reaching 96.6%. The detection efficiency of the remaining targets ranged from 66.7% to 96.0%. Various new detection methods, like Serum specific antibody detection, can speed up the test time. However, due to the complexity of the method and higher testing requirements, it seems that it cannot be used as a complete replacement for qRT-PRC testing. CONCLUSIONS With the advancement of technology and the improvement of methods, the detection methods of SARSCoV-2 have become more mature. These advances provided great help to the detection of SARS-CoV-2.
Collapse
Affiliation(s)
- Hao-jia Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000 China
| | - Yang-hui Xiang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000 China
| | - Rui Hu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000 China
| | - Rui Ji
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000 China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000 China
| | - Yu-ping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000 China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000 China
| |
Collapse
|
17
|
Suzuki K, Levert A, Yeung J, Starr M, Cameron J, Williams R, Rismanto N, Stark T, Druery D, Prasad S, Ferrarini C, Hanafi I, McNally LP, Cunningham P, Liu Z, Ishida T, Huang CS, Oswald V, Evans L, Symonds G, Brew BJ, Zaunders J. HIV-1 viral blips are associated with repeated and increasingly high levels of cell-associated HIV-1 RNA transcriptional activity. AIDS 2021; 35:2095-2103. [PMID: 34148986 PMCID: PMC8505147 DOI: 10.1097/qad.0000000000003001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Some HIV+ patients, virally suppressed on ART, show occasional 'blips' of detectable HIV-1 plasma RNA. We used a new highly sensitive assay of cell-associated HIV-1 RNA to measure transcriptional activity in PBMCs and production of infectious virus from the viral reservoir, in patients with and without 'blips'. DESIGN/METHODS RNA and DNA extracted from cells in 6 ml of peripheral blood, from suppressed patients with one to two 'blip' episodes over the past 2 years of ART (n = 55), or no 'blips' (n = 52), were assayed for HIV-1 RNA transcripts and proviral DNA targeting the highly conserved 'R' region of the LTR. Follow-up samples were also collected. Purified CD4+ T cells were cultured with anti-CD3/CD28/CD2 T-cell activator to amplify transcription and measure replication competent virus. RESULTS HIV-1 RNA transcripts ranged from 1.3 to 5415 copies/106 white blood cells. 'Blip' patients had significantly higher levels vs. without blips (median 192 vs. 49; P = 0.0007), which correlated with: higher levels of inducible transcripts after activation in vitro, sustained higher HIV-1 transcription levels in follow-up samples along with increasing HIV-1 DNA in some, and production of replication-competent HIV-1. CONCLUSION Viral 'blips' are significant reflecting higher transcriptional activity from the reservoir and contribute to the reservoir over time. This sensitive assay can be used in monitoring the size and activity of the HIV-1 reservoir and will be useful in HIV-1 cure strategies.
Collapse
Affiliation(s)
- Kazuo Suzuki
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Angelique Levert
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Julie Yeung
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Mitchell Starr
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Jane Cameron
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Raffaella Williams
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Nikolas Rismanto
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Tayla Stark
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Dylan Druery
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Salzeena Prasad
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Cristina Ferrarini
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Imelda Hanafi
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Leon Patrick McNally
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Philip Cunningham
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| | - Zhixin Liu
- Stats Central, University of New South Wales, Sydney, NSW, Australia
| | | | | | - Velma Oswald
- Clinical Immunology and HIV Medicine, Liverpool Hospital
| | - Louise Evans
- Clinical Immunology and HIV Medicine, Liverpool Hospital
- University of New South Wales
| | | | - Bruce James Brew
- Departments of Neurology and Immunology
- Peter Duncan Neurosciences Unit, St Vincent's Centre for Applied Medical Research, and University of Notre Dame
- Department of HIV Medicine, St Vincent's Hospital
- St Vincent's Clinical School, Delacy Building, University of New South Wales, Sydney, NSW, Australia
| | - John Zaunders
- St Vincent's Centre for Applied Medical Research, NSW State Reference laboratory for HIV
| |
Collapse
|
18
|
Shen J, Zheng J, Li Z, Liu Y, Jing F, Wan X, Yamaguchi Y, Zhuang S. A rapid nucleic acid concentration measurement system with large field of view for a droplet digital PCR microfluidic chip. LAB ON A CHIP 2021; 21:3742-3747. [PMID: 34378610 DOI: 10.1039/d1lc00532d] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Droplet digital polymerase chain reaction (ddPCR) is an effective technique, with unparalleled sensitivity, for the absolute quantification of target nucleic acids. However, current commercial ddPCR devices for detecting the gene chip are time consuming due to complex image stitching. To address this issue, we propose a universal concentration determination system and realize one-time gene chip imaging with high resolution. All the functional units are controlled by self-developed software using the PyQt5 module in Python. Without stitching technology, images of the ddPCR chip (28 mm × 18 mm) containing 20 000 independent 0.81 nL micro chambers can be obtained in less than 15 seconds, which saves about 165 seconds. A white laser light source (2 mW cm-2) was employed as a substitute for the mercury lamp. Its wavelength matches well with typical fluorescent dyes (e.g., HEX, ROX and Cy5), and thus it can strengthen the fluorescence intensity for weak signals. The results also demonstrated that the correlation coefficient for the measured concentration and theoretical value was above 99%, by testing the ddPCR products with COVID-19 virus. Such a system can greatly reduce the time required for image acquisition and DNA concentration determination, and thus is able to speed up the lab-to-application process for ddPCR technology.
Collapse
Affiliation(s)
- Jinrong Shen
- Shanghai Key Lab of Modern Optical System, School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Jihong Zheng
- Shanghai Key Lab of Modern Optical System, School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Zhenqing Li
- Shanghai Key Lab of Modern Optical System, School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Yourong Liu
- Shanghai Key Lab of Modern Optical System, School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Fengxiang Jing
- Shanghai Turtle Technology Limited, Shanghai 200439, China
| | - Xinjun Wan
- Shanghai Key Lab of Modern Optical System, School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Yoshinori Yamaguchi
- Oono Joint Research laboratory, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Songlin Zhuang
- Shanghai Key Lab of Modern Optical System, School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| |
Collapse
|
19
|
Ebola Virus Disease, Diagnostics and Therapeutics: Where is the Consensus in Over Three Decades of Clinical Research? SCIENTIFIC AFRICAN 2021. [DOI: 10.1016/j.sciaf.2021.e00862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
20
|
Abstract
PURPOSE OF REVIEW Despite decades of suppressive antiretroviral therapy (ART), HIV-1 reservoirs persist and fuel viral rebound if therapy is interrupted. The persistence of viral reservoirs in infected individuals is the main obstacle to achieving HIV-1 eradication or a long-term remission. Accurate assessment of the viral reservoir size is necessary for monitoring the effectiveness of the curative interventions. Here, we review the recent progress in the development of assays to measure HIV-1 persistence, highlighting their key advantages and limitations. RECENT FINDINGS To estimate the viral reservoir size, a number of assays have been developed that assess different aspects of HIV-1 persistence in ART-treated individuals. These include viral outgrowth assays to measure proviral replication competence, sequencing-based assays to measure genetic intactness of HIV-1 proviruses, and diverse techniques that measure the ability of proviruses to produce viral RNA and/or proteins (transcription and translation competence), with or without ex vivo stimulation. Recent years have seen the development of next-generation reservoir assays that, in addition to measuring viral persistence markers, assess the proviral integration sites and characterize the HIV-1 reservoir cells on the single-cell level. SUMMARY Although no assay yet can measure the HIV-1 reservoir with 100% accuracy, recent technical advances allow reliable estimation of its size and composition.
Collapse
|
21
|
Thalinger B, Pütz Y, Traugott M. Endpoint PCR coupled with capillary electrophoresis (celPCR) provides sensitive and quantitative measures of environmental DNA in singleplex and multiplex reactions. PLoS One 2021; 16:e0254356. [PMID: 34297715 PMCID: PMC8301609 DOI: 10.1371/journal.pone.0254356] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 06/24/2021] [Indexed: 11/18/2022] Open
Abstract
The use of sensitive methods is key for the detection of target taxa from trace amounts of environmental DNA (eDNA) in a sample. In this context, digital PCR (dPCR) enables direct quantification and is commonly perceived as more sensitive than endpoint PCR. However, endpoint PCR coupled with capillary electrophoresis (celPCR) potentially embodies a viable alternative as it quantitatively measures signal strength after PCR in Relative Fluorescence Units (RFU). Provided comparable levels of sensitivity are reached, celPCR permits the development of cost-efficient multiplex reactions, enabling the simultaneous detection of several target taxa. Here, we compared the sensitivity of singleplex and multiplex celPCR to dPCR for species-specific primer pairs amplifying mitochondrial DNA (COI) of fish species occurring in European freshwaters by analyzing dilution series of tissue extracts as well as field-collected water samples. Both singleplex and multiplex celPCR and dPCR displayed comparable sensitivity with reliable positive amplifications starting at two to 10 target DNA copies per μl extract. celPCR was suitable for quantifying target DNA and direct inference of copy numbers from RFU was possible after accounting for primer effects in linear mixed-effects models and calibration via dPCR. Furthermore, multiplex celPCR and dPCR were successfully used for the detection and quantification of fish-eDNA in field-collected water samples, confirming the results of the dilution series experiment and exemplifying the high sensitivity of the two approaches. The possibility of detection and quantification via multiplex celPCR is appealing for the cost-efficient screening of high sample numbers. The present results confirm the sensitivity of this approach thus enabling its application for future eDNA-based monitoring efforts.
Collapse
Affiliation(s)
- Bettina Thalinger
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
- Centre for Biodiversity Genomics, University of Guelph, Guelph, Ontario, Canada
- Department of Integrative Biology, College of Biological Science, University of Guelph, Guelph, Ontario, Canada
| | - Yannick Pütz
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Michael Traugott
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
- Sinsoma GmbH, Lannes 6, Völs, Austria
| |
Collapse
|
22
|
Kojabad AA, Farzanehpour M, Galeh HEG, Dorostkar R, Jafarpour A, Bolandian M, Nodooshan MM. Droplet digital PCR of viral DNA/RNA, current progress, challenges, and future perspectives. J Med Virol 2021; 93:4182-4197. [PMID: 33538349 PMCID: PMC8013307 DOI: 10.1002/jmv.26846] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/01/2021] [Indexed: 12/18/2022]
Abstract
High-throughput droplet-based digital PCR (ddPCR) is a refinement of the conventional polymerase chain reaction (PCR) methods. In ddPCR, DNA/RNA is encapsulated stochastically inside the microdroplets as reaction chambers. A small percentage of the reaction chamber contains one or fewer copies of the DNA or RNA. After PCR amplification, concentrations are determined based on the proportion of nonfluorescent partitions through the Poisson distribution. Some of the main features of ddPCR include high sensitivity and specificity, absolute quantification without a standard curve, high reproducibility, good tolerance to PCR inhibitor, and high efficacy compared to conventional molecular methods. These advantages make ddPCR a valuable addition to the virologist's toolbox. The following review outlines the recent technological advances in ddPCR methods and their applications in viral identification.
Collapse
Affiliation(s)
- Amir Asri Kojabad
- Applied Virology Research CenterBaqiyatallah University of Medical SciencesTehranIran
| | - Mahdieh Farzanehpour
- Applied Virology Research CenterBaqiyatallah University of Medical SciencesTehranIran
| | | | - Ruhollah Dorostkar
- Applied Virology Research CenterBaqiyatallah University of Medical SciencesTehranIran
| | - Ali Jafarpour
- Research Center for Clinical VirologyTehran University of Medical SciencesTehranIran
| | - Masoumeh Bolandian
- Applied Virology Research CenterBaqiyatallah University of Medical SciencesTehranIran
| | | |
Collapse
|
23
|
Kuhn L, Paximadis M, Da Costa Dias B, Shen Y, Mncube S, Strehlau R, Shiau S, Patel F, Burke M, Technau KG, Sherman G, Loubser S, Abrams EJ, Tiemessen CT. Predictors of cell-associated HIV-1 DNA over one year in very early treated infants. Clin Infect Dis 2021; 74:1047-1054. [PMID: 34185838 DOI: 10.1093/cid/ciab586] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Younger age of antiretroviral therapy (ART) initiation is associated with a smaller viral reservoir size in perinatally-acquired HIV-1 infection, but there is wide variability among early-treated infants. Predictors of this variability are not fully described. METHODS Sixty-three neonates diagnosed with HIV-1 <48 hours after birth in Johannesburg, South Africa were started on ART as soon as possible. Fifty-nine (94%) infants received daily nevirapine prophylaxis from birth until ART start. Viably-preserved peripheral blood mononuclear cells (PBMC) collected at regular intervals to 48 weeks, and from respective mothers at enrolment, were tested using integrase-targeted, semi-nested, real-time quantitative hydrolysis probe (TaqMan) PCR assays to quantify total HIV-1 subtype C viral DNA (vDNA). Predictors were investigated using Generalized Estimating Equation (GEE) regression models. RESULTS Thirty-one (49.2%) infants initiated ART <48 hours, 24 (38.1%) <14 days and 8 (12.7%) >14 days of birth. Three-quarters were infected despite maternal antenatal ART (however, only 9.5% of women had undetectable viral load closest to delivery) and 86% were breastfed. Higher infant CD4+ T-cell percentage and viral load <100,000 copies/ml pre-ART were associated with lower levels of vDNA copies/10 6 PBMC equivalents in the first 48 weeks after ART start. No antenatal maternal ART and breastfeeding were also associated with lower vDNA. Older age at ART initiation had a discernible negative impact when initiated >14 days. CONCLUSIONS Among very early treated infants, higher CD4+ T-cell percentage and viral load <100,000 copies/ml pre-ART, infection occurring in the absence of maternal antenatal ART and breastfeeding were associated with lower levels of HIV-1 DNA in the first 48 weeks of treatment.
Collapse
Affiliation(s)
- Louise Kuhn
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| | - Maria Paximadis
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services, and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Bianca Da Costa Dias
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services, and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Yanhan Shen
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Sizanani Mncube
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services, and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Renate Strehlau
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stephanie Shiau
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ
| | - Faeezah Patel
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Wits Reproductive Health and HIV Institute (WRHI), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Megan Burke
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Karl-Günter Technau
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gayle Sherman
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services, and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shayne Loubser
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services, and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Elaine J Abrams
- ICAP at Columbia University, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA.,Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Caroline T Tiemessen
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services, and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | |
Collapse
|
24
|
Katusiime MG, Van Zyl GU, Cotton MF, Kearney MF. HIV-1 Persistence in Children during Suppressive ART. Viruses 2021; 13:v13061134. [PMID: 34204740 PMCID: PMC8231535 DOI: 10.3390/v13061134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 12/16/2022] Open
Abstract
There is a growing number of perinatally HIV-1-infected children worldwide who must maintain life-long ART. In early life, HIV-1 infection is established in an immunologically inexperienced environment in which maternal ART and immune dynamics during pregnancy play a role in reservoir establishment. Children that initiated early antiretroviral therapy (ART) and maintained long-term suppression of viremia have smaller and less diverse HIV reservoirs than adults, although their proviral landscape during ART is reported to be similar to that of adults. The ability of these early infected cells to persist long-term through clonal expansion poses a major barrier to finding a cure. Furthermore, the effects of life-long HIV persistence and ART are yet to be understood, but growing evidence suggests that these individuals are at an increased risk for developing non-AIDS-related comorbidities, which underscores the need for an HIV cure.
Collapse
Affiliation(s)
- Mary Grace Katusiime
- HIV Dynamics and Replication Program, CCR, National Cancer Institute, Frederick, MD 21702, USA;
- Correspondence:
| | - Gert U. Van Zyl
- Division of Medical Virology, Stellenbosch University and National Health Laboratory Service Tygerberg, Cape Town 8000, South Africa;
| | - Mark F. Cotton
- Department of Pediatrics and Child Health, Tygerberg Children’s Hospital and Family Center for Research with Ubuntu, Stellenbosch University, Cape Town 7505, South Africa;
| | - Mary F. Kearney
- HIV Dynamics and Replication Program, CCR, National Cancer Institute, Frederick, MD 21702, USA;
| |
Collapse
|
25
|
Kiulia NM, Gonzalez R, Thompson H, Aw TG, Rose JB. Quantification and Trends of Rotavirus and Enterovirus in Untreated Sewage Using Reverse Transcription Droplet Digital PCR. FOOD AND ENVIRONMENTAL VIROLOGY 2021; 13:154-169. [PMID: 33591485 DOI: 10.1007/s12560-020-09455-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 12/04/2020] [Indexed: 06/12/2023]
Abstract
The quantification and trends in concentrations for naturally occurring rotaviruses (RV) and enteroviruses (EV) in untreated sewage in various wastewater systems have not often been compared. There is now greater interest in monitoring the infections in the community including live vaccine efficacy by evaluating untreated sewage. The goals of this study were to 1) survey the concentrations of naturally occurring RV and EV in untreated sewage using a reverse transcription-droplet digital polymerase chain reaction (RT-ddPCR) and 2) investigate the use of a new adsorption elution (bag-mediated filtration system (BMFS) using ViroCap filters) against more traditional polyethylene glycol (PEG) precipitation for virus concentration. Sewage samples were collected from lagoons in Kenya and Michigan (MI), the United States (USA) and from wastewater treatment plants (WWTPs) in the USA. RVs were detected at geometric mean concentrations in various locations, California (CA) 1.31 × 105 genome copies/L (gc/L), Kenya (KE) 2.71 × 104 gc/L and Virginia (VA) 1.48 × 105 gc/L, and EVs geometric means were 3.72 × 106 gc/L (CA), 1.18 × 104 gc/L (Kenya), and 6.18 × 103 gc/L (VA). The mean RV concentrations using BMFS-ViroCap in split samples compared to PEG precipitation methods demonstrated that the levels were only 9% (#s BMFS/PEG) in the Michigan lagoons which was significantly different (p < 0.01). This suggests that RV concentrations in Kenya are around 1.69 × 106 gc/L. Overall, there was no difference in concentrations for the other sampling locations across the methods of virus recovery (i.e., PEG precipitation and HA filters) using one-way ANOVA (F = 1.7, p = 0.2739) or Tukey-Kramer pairwise comparisons (p > 0.05). This study provides useful data on RV and EV concentrations in untreated sewage in Kenya and the USA. It also highlights on the usefulness of the RT-ddPCR for absolute quantification of RV and EV in sewage samples. The BMFS using ViroCap filters while less efficient compared to the more traditional PEG precipitation method was able to recover RVs and EVs in untreated sewage and may be useful in poor resource settings while underestimating viruses by 1 to 1.5 logs.
Collapse
Affiliation(s)
- Nicholas M Kiulia
- The Water Quality, Environmental and Molecular Microbiology Laboratory, Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI, 48824, USA.
- Enteric Pathogens and Water Research Laboratory, Institute of Primate Research, P.O. Box 24481-00502, Karen, Nairobi, Kenya.
| | - Raul Gonzalez
- Hampton Roads Sanitation District, 1434 Air Rail Avenue, Virginia Beach, VA, 23455, USA
| | - Hannah Thompson
- Hampton Roads Sanitation District, 1434 Air Rail Avenue, Virginia Beach, VA, 23455, USA
| | - Tiong Gim Aw
- Department of Environmental Health Sciences, Tulane University, 1440 Canal Street, Suite 2100, New Orleans, LA, 70112, USA
| | - Joan B Rose
- The Water Quality, Environmental and Molecular Microbiology Laboratory, Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
26
|
Abstract
Droplet digital polymerase chain reaction (ddPCR) is a method used to detect and quantify nucleic acids even when present in exceptionally low numbers. While it has proven to be valuable for clinical studies, it has failed to be widely adopted for environmental studies but despite some limitations, ddPCR may represent a better option than classical qPCR for environmental samples. Due to the complexity of the chemical and biological composition of environmental samples, protocols tailored to clinical studies are not appropriate, and results are difficult to interpret. We used environmental DNA samples originating from field studies to determine a protocol for environmental samples. Samples included field soils which had been inoculated with the soil fungus Rhizophagus irregularis (environmental positive control), field soils that had not been inoculated and the targeted fungus was not naturally present (environmental negative control), and root samples from both field categories. To control for the effect of soil inhibitors, we also included DNA samples of an organismal control extracted from pure fungal spores (organismal positive control). Finally, we included a no-template control consisting only of the PCR reaction reagents and nuclease free water instead of template DNA. Using original data, we examined which factors contribute to poor resolution in root and soil samples and propose best practices to ensure accuracy and repeatability. Furthermore, we evaluated manual and automatic threshold determination methods and we propose a novel protocol based on multiple controls that is more appropriate for environmental samples.
Collapse
|
27
|
Tytgat O, Tang MX, van Snippenberg W, Boel A, Guggilla RR, Gansemans Y, Van Herp M, Symoens S, Trypsteen W, Deforce D, Heindryckx B, Coucke P, De Spiegelaere W, Van Nieuwerburgh F. Digital Polymerase Chain Reaction for Assessment of Mutant Mitochondrial Carry-over after Nuclear Transfer for In Vitro Fertilization. Clin Chem 2021; 67:968-976. [PMID: 33822904 DOI: 10.1093/clinchem/hvab021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/21/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND The quantification of mitochondrial DNA heteroplasmy for the diagnosis of mitochondrial disease or after mitochondrial donation, is performed mainly using next-generation sequencing strategies (NGS). Digital PCR (dPCR) has the potential to offer an accurate alternative for mutation load quantification. METHODS We assessed the mutation load of 23 low-input human samples at the m.11778 locus, which is associated with Leber's hereditary optic neuropathy (LHON) using 2 droplet digital PCR platforms (Stilla Naica and Bio-Rad QX200) and the standard NGS strategy. Assay validation was performed by analyzing a titration series with mutation loads ranging from 50% to 0.01%. RESULTS A good concordance in mutation rates was observed between both dPCR techniques and NGS. dPCR established a distinctly lower level of background noise compared to NGS. Minor alleles with mutation loads lower than 1% could still be detected, with standard deviations of the technical replicates varying between 0.07% and 0.44% mutation load. Although no significant systematic bias was observed when comparing dPCR and NGS, a minor proportional bias was detected. A slight overestimation of the minor allele was observed for the NGS data, most probably due to amplification and sequencing errors in the NGS workflow. CONCLUSION dPCR has proven to be an accurate tool for the quantification of mitochondrial heteroplasmy, even for samples harboring a low mutation load (<1%). In addition, this alternative technique holds multiple benefits compared to NGS (e.g., less hands-on time, more straightforward data-analysis, and a lower up-front capital investment).
Collapse
Affiliation(s)
- Olivier Tytgat
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium.,Department of Life Science Technologies, Imec, Leuven, Belgium
| | - Mao-Xing Tang
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - Willem van Snippenberg
- Department of Internal Medicine and Pediatrics, HIV Cure Research Center, Ghent University, Ghent, Belgium
| | - Annekatrien Boel
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ramesh Reddy Guggilla
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - Yannick Gansemans
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | - Michiel Van Herp
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Sofie Symoens
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent (CMGG), Ghent University Hospital, Ghent, Belgium
| | - Wim Trypsteen
- Department of Internal Medicine and Pediatrics, HIV Cure Research Center, Ghent University, Ghent, Belgium
| | - Dieter Deforce
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | - Björn Heindryckx
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - Paul Coucke
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent (CMGG), Ghent University Hospital, Ghent, Belgium
| | - Ward De Spiegelaere
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | |
Collapse
|
28
|
Huggett JF. The Digital MIQE Guidelines Update: Minimum Information for Publication of Quantitative Digital PCR Experiments for 2020. Clin Chem 2021; 66:1012-1029. [PMID: 32746458 DOI: 10.1093/clinchem/hvaa125] [Citation(s) in RCA: 224] [Impact Index Per Article: 74.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/18/2020] [Indexed: 12/17/2022]
Abstract
Digital PCR (dPCR) has developed considerably since the publication of the Minimum Information for Publication of Digital PCR Experiments (dMIQE) guidelines in 2013, with advances in instrumentation, software, applications, and our understanding of its technological potential. Yet these developments also have associated challenges; data analysis steps, including threshold setting, can be difficult and preanalytical steps required to purify, concentrate, and modify nucleic acids can lead to measurement error. To assist independent corroboration of conclusions, comprehensive disclosure of all relevant experimental details is required. To support the community and reflect the growing use of dPCR, we present an update to dMIQE, dMIQE2020, including a simplified dMIQE table format to assist researchers in providing key experimental information and understanding of the associated experimental process. Adoption of dMIQE2020 by the scientific community will assist in standardizing experimental protocols, maximize efficient utilization of resources, and further enhance the impact of this powerful technology.
Collapse
|
29
|
Park C, Lee J, Hassan ZU, Ku KB, Kim SJ, Kim HG, Park EC, Park GS, Park D, Baek SH, Park D, Lee J, Jeon S, Kim S, Lee CS, Yoo HM, Kim S. Comparison of Digital PCR and Quantitative PCR with Various SARS-CoV-2 Primer-Probe Sets. J Microbiol Biotechnol 2021; 31:358-367. [PMID: 33397829 PMCID: PMC9705847 DOI: 10.4014/jmb.2009.09006] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/09/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022]
Abstract
The World Health Organization (WHO) has declared the coronavirus disease 2019 (COVID-19) as an international health emergency. Current diagnostic tests are based on the reverse transcription-quantitative polymerase chain reaction (RT-qPCR) method, which is the gold standard test that involves the amplification of viral RNA. However, the RT-qPCR assay has limitations in terms of sensitivity and quantification. In this study, we tested both qPCR and droplet digital PCR (ddPCR) to detect low amounts of viral RNA. The cycle threshold (CT) of the viral RNA by RT-PCR significantly varied according to the sequences of the primer and probe sets with in vitro transcript (IVT) RNA or viral RNA as templates, whereas the copy number of the viral RNA by ddPCR was effectively quantified with IVT RNA, cultured viral RNA, and RNA from clinical samples. Furthermore, the clinical samples were assayed via both methods, and the sensitivity of the ddPCR was determined to be equal to or more than that of the RT-qPCR. However, the ddPCR assay is more suitable for determining the copy number of reference materials. These findings suggest that the qPCR assay with the ddPCR defined reference materials could be used as a highly sensitive and compatible diagnostic method for viral RNA detection.
Collapse
Affiliation(s)
- Changwoo Park
- Microbiological Analysis Team, Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea,National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea,Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Jina Lee
- Microbiological Analysis Team, Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea,College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Zohaib ul Hassan
- Microbiological Analysis Team, Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea,Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea,Department of Bio-Analytical Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Keun Bon Ku
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Seong-Jun Kim
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Hong Gi Kim
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Edmond Changkyun Park
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea,Department of Bio-Analytical Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea,Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Gun-Soo Park
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea,Research Group of Food Processing, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
| | - Daeui Park
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea,Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Seung-Hwa Baek
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea,Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Dongju Park
- Microbiological Analysis Team, Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea,Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea,Department of Biological Science, Chungnam National University College of Bioscience and Biotechnology, Daejeon 34134, Republic of Korea
| | - Jihye Lee
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Sangeun Jeon
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Seungtaek Kim
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Chang-Seop Lee
- Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju 54986, Republic of Korea,Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| | - Hee Min Yoo
- Microbiological Analysis Team, Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea,Department of Bio-Analytical Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea,Corresponding authors H.M. Yoo Phone/Fax: +82-42-868-5362 E-mail:
| | - Seil Kim
- Microbiological Analysis Team, Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea,Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea,Department of Bio-Analytical Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea,S. Kim Phone: +82-42-868-5619 Fax: +82-42-868-5801 E-mail:
| |
Collapse
|
30
|
Duong K, Ou J, Li Z, Lv Z, Dong H, Hu T, Zhang Y, Hanna A, Gordon S, Crynen G, Head SR, Ordoukhanian P, Wang Y. Increased sensitivity using real-time dPCR for detection of SARS-CoV-2. Biotechniques 2021; 70:7-20. [PMID: 33222514 PMCID: PMC7888512 DOI: 10.2144/btn-2020-0133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
A real-time dPCR system was developed to improve the sensitivity, specificity and quantification accuracy of end point dPCR. We compared three technologies - real-time qPCR, end point dPCR and real-time dPCR - in the context of SARS-CoV-2. Some improvement in limit of detection was obtained with end point dPCR compared with real-time qPCR, and the limit of detection was further improved with the newly developed real-time dPCR technology through removal of false-positive signals. Real-time dPCR showed increased linear dynamic range compared with end point dPCR based on quantitation from amplification curves. Real-time dPCR can improve the performance of TaqMan assays beyond real-time qPCR and end point dPCR with better sensitivity and specificity, absolute quantification and a wider linear range of detection.
Collapse
Affiliation(s)
- Kyra Duong
- Gnomegen, 6440 Lusk Blvd, D207, San Diego, CA 92121, USA
| | - Jiajia Ou
- QuestGenomics, 12 E. Mozhou Rd, U-Park, Rm P308, Jiangning, Nanjing, Jiangsu, PR China
| | - Zhaoliang Li
- QuestGenomics, 12 E. Mozhou Rd, U-Park, Rm P308, Jiangning, Nanjing, Jiangsu, PR China
| | - Zhaoqing Lv
- QuestGenomics, 12 E. Mozhou Rd, U-Park, Rm P308, Jiangning, Nanjing, Jiangsu, PR China
| | - Hao Dong
- QuestGenomics, 12 E. Mozhou Rd, U-Park, Rm P308, Jiangning, Nanjing, Jiangsu, PR China
| | - Tao Hu
- QuestGenomics, 12 E. Mozhou Rd, U-Park, Rm P308, Jiangning, Nanjing, Jiangsu, PR China
| | - Yunyun Zhang
- QuestGenomics, 12 E. Mozhou Rd, U-Park, Rm P308, Jiangning, Nanjing, Jiangsu, PR China
| | - Ava Hanna
- Gnomegen, 6440 Lusk Blvd, D207, San Diego, CA 92121, USA
| | - Skyler Gordon
- Genomics Core Facility, Scripps Research, 10550 N Torrey Pines Road, La Jolla, CA, USA
| | - Gogce Crynen
- Bioinformatics Core Facility, Scripps Research, 120 Scripps Way, Jupiter, FL 33458, USA
| | - Steven R Head
- Genomics Core Facility, Scripps Research, 10550 N Torrey Pines Road, La Jolla, CA, USA
| | - Phillip Ordoukhanian
- Genomics Core Facility, Scripps Research, 10550 N Torrey Pines Road, La Jolla, CA, USA
| | - Yan Wang
- Gnomegen, 6440 Lusk Blvd, D207, San Diego, CA 92121, USA
| |
Collapse
|
31
|
Tellinghuisen J. dPCR vs. qPCR: The role of Poisson statistics at low concentrations. Anal Biochem 2020; 611:113946. [DOI: 10.1016/j.ab.2020.113946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 08/14/2020] [Accepted: 09/03/2020] [Indexed: 10/23/2022]
|
32
|
Xu G, Si H, Jing F, Sun P, Zhao D, Wu D. A Double-Deck Self-Digitization Microfluidic Chip for Digital PCR. MICROMACHINES 2020; 11:mi11121025. [PMID: 33255151 PMCID: PMC7759810 DOI: 10.3390/mi11121025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/31/2022]
Abstract
In this work, a double-deck microfluidic chip was presented for digital PCR application. This chip consists of two reverse-placed micro-patterned polydimethylsiloxane (PDMS) layers between the top and bottom glass substrates. Each micropatterned PDMS layer contains more than 20,000 cylindrical micro-chambers to hold the partitioned droplets. The double-deck designs can double the number of chambers and reagent capacity without changing the planar area of the chip. In addition, carbon black was mixed into the pure PDMS gel to obstruct the passage of fluorescence from the positive chambers between the two layers of the chip. Thus, the fluorescence signal of micro-chambers in different layers of the chip after PCR can be collected without mutual interference. The quantitative capability of the proposed chip was evaluated by measuring a 10-fold serial dilution of the DNA template. A high accuracy of the absolute quantification for nucleic acid with a dynamic range of 105 was demonstrated by this chip in this work. Owing to its characteristics of small planar area, large capacity, and sensitivity, the double-deck microfluidic chip is expected to further promote the extensive applications of digital PCR.
Collapse
Affiliation(s)
- Gangwei Xu
- State Key Laboratory of ASIC and System, School of Microelectronics, Fudan University, Shanghai 200433, China; (G.X.); (H.S.); (P.S.); (D.Z.)
| | - Huaqing Si
- State Key Laboratory of ASIC and System, School of Microelectronics, Fudan University, Shanghai 200433, China; (G.X.); (H.S.); (P.S.); (D.Z.)
| | - Fengxiang Jing
- Turtle Technology Company Limited, Shanghai 200439, China;
| | - Peng Sun
- State Key Laboratory of ASIC and System, School of Microelectronics, Fudan University, Shanghai 200433, China; (G.X.); (H.S.); (P.S.); (D.Z.)
| | - Dan Zhao
- State Key Laboratory of ASIC and System, School of Microelectronics, Fudan University, Shanghai 200433, China; (G.X.); (H.S.); (P.S.); (D.Z.)
| | - Dongping Wu
- State Key Laboratory of ASIC and System, School of Microelectronics, Fudan University, Shanghai 200433, China; (G.X.); (H.S.); (P.S.); (D.Z.)
- Correspondence:
| |
Collapse
|
33
|
Chen M, Wu D, Tu S, Yang C, Chen D, Xu Y. CRISPR/Cas9 cleavage triggered ESDR for circulating tumor DNA detection based on a 3D graphene/AuPtPd nanoflower biosensor. Biosens Bioelectron 2020; 173:112821. [PMID: 33221510 DOI: 10.1016/j.bios.2020.112821] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 12/15/2022]
Abstract
Circulating tumor DNA (ctDNA) plays an important role in the early diagnosis and prognosis of several cancers and is a credible biomarker for predicting the response to therapy. Additionally, the fact that the strategy used to detect ctDNA is non-invasive also adds to the advantages of using ctDNA for predicting disease diagnosis and prognosis. However, low abundance in peripheral blood and the high background of wild-type DNA impair the precise and specific measurement of ctDNA. In this study, we developed a novel 3D GR/AuPtPd nanoflower sensing platform based on CRISPR/Cas9 cleavage-triggered entropy-driven strand displacement reaction (ESDR) for the effective detection of ctDNA. Low levels of ctDNA could be detected using this method as the ESDR amplification does require complicated operation procedures and stringent reaction conditions. By combining the advantages of the site-specific cleavage by "gene magic scissors," Cas9/sgRNA, with those of the rapid amplification kinetics of entropy-driven strand displacement, our method resulted in amplification efficiency as well as high specificity for discriminating single-nucleotide mismatches. The 3D GR/AuPtPd nanoflower-based electrochemical biosensor displayed high specificity and worthy performance in assays with human serum. Therefore, this pioneered method provides a new paradigm for efficient ctDNA detection and shows great potential for use in clinical and diagnostic applications.
Collapse
Affiliation(s)
- Mei Chen
- Clinical Laboratory, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Dongming Wu
- Clinical Laboratory, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Shihua Tu
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Chaoyin Yang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - DeJie Chen
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Ying Xu
- Clinical Laboratory, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China.
| |
Collapse
|
34
|
Kathrada AI, Wei SC, Xu Y, Cheow, LF, Chen CH. Microfluidic compartmentalization to identify gene biomarkers of infection. BIOMICROFLUIDICS 2020; 14:061502. [PMID: 33312326 PMCID: PMC7717927 DOI: 10.1063/5.0032849] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 11/09/2020] [Indexed: 05/20/2023]
Abstract
Infectious diseases caused by pathogens, such as SARS-COV, H7N9, severe fever with thrombocytopenia syndrome virus, and human immunodeficiency virus, have fatal outcomes with common features of severe fever and subsequent bacterial invasion progressing to multiorgan failure. Gene biomarkers are promising to distinguish specific infections from others with similar presenting symptoms for the prescription of correct therapeutics, preventing pandemics. While routine laboratory methods based on polymerase chain reaction (PCR) to measure gene biomarkers have provided highly sensitive and specific viral detection techniques over the years, they are still hampered by their precision and resource intensity precluding their point-of-care use. Recently, there has been growing interest in employing microfluidic technologies to advance current methods for infectious disease determination via gene biomarker measurements. Here, based on the requirement of infection detection, we will review three microfluidic approaches to compartmentalize gene biomarkers: (1) microwell-based PCR platforms; (2) droplet-based PCR; and (3) point-of-care devices including centrifugal chip, SlipChip, and self-powered integrated microfluidic point-of-care low-cost enabling chip. By capturing target genes in microwells with a small sample volume (∼μl), sensitivity can be enhanced. Additionally, with the advance of significant sample volume minimization (∼pl) using droplet technology, gene quantification is possible. These improvements in cost, automation, usability, and portability have thereby allowed point-of-care applications to decentralize testing platforms from laboratory-based settings to field use against infections.
Collapse
Affiliation(s)
- Ahmad Ismat Kathrada
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Block 4, #04-08, Singapore 117583
| | | | - Ying Xu
- Department of Biomedical Engineering, City University of Hong Kong, Room Y6700, 6/F, Yeung Kin Man Academic Building, 83 Tat Chee Avenue, Hong Kong, China
| | | | - Chia-Hung Chen
- Department of Biomedical Engineering, City University of Hong Kong, Room Y6700, 6/F, Yeung Kin Man Academic Building, 83 Tat Chee Avenue, Hong Kong, China
- Author to whom correspondence should be addressed:
| |
Collapse
|
35
|
Development of droplet digital PCR for the detection of Tilletia laevis, which causes common bunt of wheat, based on the SCAR marker derived from ISSR and real-time PCR. Sci Rep 2020; 10:16106. [PMID: 32999395 PMCID: PMC7528053 DOI: 10.1038/s41598-020-72976-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/28/2020] [Indexed: 01/15/2023] Open
Abstract
Common bunt of wheat caused by Tilletia laevis and/or T. caries (syn. T. tritici), is a major disease in wheat-growing regions worldwide that could lead to 80% or even total loss of production. Even though T. laevis can be distinguished from T. caries on the bases of morphology of teliospores using microscopy technique. However, molecular methods could serve as an additional method to quantify the pathogen. To develop a rapid diagnostic and quantify method, we employed the ISSR molecular marker for T. laevis in this study. The primer ISSR857 generated a polymorphic pattern displaying a 1385 bp T. laevis-specific DNA fragment. A pair of specific primers (L57F/L57R) was designed to amplify a sequence-characterized amplified region (SCAR) (763 bp) for the PCR detection assay. The primers amplified the DNA fragment in the tested isolates of T. laevis but failed in the related species, including T. caries. The detection limit of the primer set (L57F/L57R) was 5 ng/µl of DNA extracted from T. laevis teliospores. A SYBR Green I real-time PCR method for detecting T. laevis with a 100 fg/µl detection limit and droplet digital PCR with a high sensitivity (30 fg/µl detection limit) were developed; this technique showed the most sensitive detection compared to the SCAR marker and SYBR Green I real-time PCR. Additionally, this is the first study related the detection of T. laevis with the droplet digital PCR method.
Collapse
|
36
|
Quantitative Detection of Beef and Beef Meat Products Adulteration by the Addition of Duck Meat Using Micro Drop Digital Polymerase Chain Reaction. J FOOD QUALITY 2020. [DOI: 10.1155/2020/2843056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A single-copy specific primer was designed based on beef and duck samples and through drop digital polymerase chain reaction (ddPCR) for the quantitative analysis. Results revealed that the primers had no specific amplification with sheep, chicken, pork, or other species. Both the relationships between meat weight and DNA weight and between DNA weight and DNA copy number (C) were nearly linear within the dynamic range. To calculate the original meat weight from the DNA copy number, the DNA weight was used as the intermediate value to establish the following formulae: Mbeef = 0.058C − 1.86; Mduck = 0.0268C − 7.78. To achieve a good quantitative analysis, all species used in the experiment were made of lean meat. The accuracy of the method was verified by artificial adulteration of different proportions. Testing of the commercial samples indicated that adulteration is present in the market. The established digital PCR method provided an effective tool for monitoring the adulterated meat products and reducing the adulteration in the market.
Collapse
|
37
|
Cao WW, He DS, Chen ZJ, Zuo YZ, Chen X, Chang YL, Zhang ZG, Ye L, Shi L. Development of a droplet digital PCR for detection and quantification of porcine epidemic diarrhea virus. J Vet Diagn Invest 2020; 32:572-576. [PMID: 32552416 DOI: 10.1177/1040638720924753] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Porcine epidemic diarrhea, a disease caused by porcine epidemic diarrhea virus (PEDV), results in large economic losses to the global swine industry. To manage this disease effectively, it is essential to detect PEDV early and accurately. We developed a sensitive and accurate droplet digital PCR (ddPCR) assay to detect PEDV. The optimal primer-to-probe concentration and melting temperature were identified as 300:200 nM and 59.2°C, respectively. The specificity of the ddPCR assay was confirmed by negative test results for common swine pathogens. The detection limit for the ddPCR was 0.26 copies/μL, which is a 5.7-fold increase in sensitivity compared to that of real-time PCR (rtPCR). Both ddPCR and rtPCR assays exhibited good linearity, although ddPCR provided higher sensitivity for clinical detection compared to that of rtPCR. Our ddPCR methodology provides a promising tool for evaluating the PEDV viral load when used for clinical testing, particularly for detecting samples with low-copy viral loads.
Collapse
Affiliation(s)
- Wei W Cao
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China (Cao, Z.J. Chen, X. Chen, Chang, Shi, Ye); College of Veterinary Medicine, South China Agricultural University, Guangzhou, China (He); College of Animal Science and Technology, Agricultural University of Hebei, Baoding, China (Zuo); State Key Laboratory of Food Safety Technology for Meat Products and Synergetic Innovation Center of Food Safety and Nutrition, Xiamen Yinxiang Group, Xiamen, China (Zhang, Shi)
| | - Dong S He
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China (Cao, Z.J. Chen, X. Chen, Chang, Shi, Ye); College of Veterinary Medicine, South China Agricultural University, Guangzhou, China (He); College of Animal Science and Technology, Agricultural University of Hebei, Baoding, China (Zuo); State Key Laboratory of Food Safety Technology for Meat Products and Synergetic Innovation Center of Food Safety and Nutrition, Xiamen Yinxiang Group, Xiamen, China (Zhang, Shi)
| | - Zhen J Chen
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China (Cao, Z.J. Chen, X. Chen, Chang, Shi, Ye); College of Veterinary Medicine, South China Agricultural University, Guangzhou, China (He); College of Animal Science and Technology, Agricultural University of Hebei, Baoding, China (Zuo); State Key Laboratory of Food Safety Technology for Meat Products and Synergetic Innovation Center of Food Safety and Nutrition, Xiamen Yinxiang Group, Xiamen, China (Zhang, Shi)
| | - Yu Z Zuo
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China (Cao, Z.J. Chen, X. Chen, Chang, Shi, Ye); College of Veterinary Medicine, South China Agricultural University, Guangzhou, China (He); College of Animal Science and Technology, Agricultural University of Hebei, Baoding, China (Zuo); State Key Laboratory of Food Safety Technology for Meat Products and Synergetic Innovation Center of Food Safety and Nutrition, Xiamen Yinxiang Group, Xiamen, China (Zhang, Shi)
| | - Xun Chen
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China (Cao, Z.J. Chen, X. Chen, Chang, Shi, Ye); College of Veterinary Medicine, South China Agricultural University, Guangzhou, China (He); College of Animal Science and Technology, Agricultural University of Hebei, Baoding, China (Zuo); State Key Laboratory of Food Safety Technology for Meat Products and Synergetic Innovation Center of Food Safety and Nutrition, Xiamen Yinxiang Group, Xiamen, China (Zhang, Shi)
| | - Yan L Chang
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China (Cao, Z.J. Chen, X. Chen, Chang, Shi, Ye); College of Veterinary Medicine, South China Agricultural University, Guangzhou, China (He); College of Animal Science and Technology, Agricultural University of Hebei, Baoding, China (Zuo); State Key Laboratory of Food Safety Technology for Meat Products and Synergetic Innovation Center of Food Safety and Nutrition, Xiamen Yinxiang Group, Xiamen, China (Zhang, Shi)
| | - Zhi G Zhang
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China (Cao, Z.J. Chen, X. Chen, Chang, Shi, Ye); College of Veterinary Medicine, South China Agricultural University, Guangzhou, China (He); College of Animal Science and Technology, Agricultural University of Hebei, Baoding, China (Zuo); State Key Laboratory of Food Safety Technology for Meat Products and Synergetic Innovation Center of Food Safety and Nutrition, Xiamen Yinxiang Group, Xiamen, China (Zhang, Shi)
| | - Lei Ye
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China (Cao, Z.J. Chen, X. Chen, Chang, Shi, Ye); College of Veterinary Medicine, South China Agricultural University, Guangzhou, China (He); College of Animal Science and Technology, Agricultural University of Hebei, Baoding, China (Zuo); State Key Laboratory of Food Safety Technology for Meat Products and Synergetic Innovation Center of Food Safety and Nutrition, Xiamen Yinxiang Group, Xiamen, China (Zhang, Shi)
| | - Lei Shi
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China (Cao, Z.J. Chen, X. Chen, Chang, Shi, Ye); College of Veterinary Medicine, South China Agricultural University, Guangzhou, China (He); College of Animal Science and Technology, Agricultural University of Hebei, Baoding, China (Zuo); State Key Laboratory of Food Safety Technology for Meat Products and Synergetic Innovation Center of Food Safety and Nutrition, Xiamen Yinxiang Group, Xiamen, China (Zhang, Shi)
| |
Collapse
|
38
|
Orlandi C, Canovari B, Bozzano F, Marras F, Pasquini Z, Barchiesi F, De Maria A, Magnani M, Casabianca A. A comparative analysis of unintegrated HIV-1 DNA measurement as a potential biomarker of the cellular reservoir in the blood of patients controlling and non-controlling viral replication. J Transl Med 2020; 18:204. [PMID: 32429953 PMCID: PMC7236182 DOI: 10.1186/s12967-020-02368-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The persistence of HIV-1 in reservoir cells is one of the major obstacles to eradicating the virus in infected individuals receiving combination antiretroviral therapy (ART). HIV-1 persists in infected cells as a stable integrated genome and more labile unintegrated DNA (uDNA), which includes linear, 1-LTR and 2-LTR circular DNA. 2-LTR circle DNA, although less abundant, is considered a surrogate marker of recent infection events and is currently used instead of the other unintegrated species as a diagnostic tool. This pilot study aimed to investigate how to best achieve the measurement of uDNA. METHODS A comparative analysis of two qPCR-based methods (U-assay and 2-LTR assay) was performed on the blood of 12 ART-naïve, 14 viremic and 29 aviremic On-ART patients and 20 untreated spontaneous controllers (HIC), sampled at a single time point. RESULTS The U-assay, which quantified all unintegrated DNA species, showed greater sensitivity than the 2-LTR assay (up to 75%, p < 0.0001), especially in viremic subjects, in whom other forms, in addition to 2-LTR circles, may also accumulate due to active viral replication. Indeed, in aviremic On-ART samples, the U-assay unexpectedly measured uDNA in a higher proportion of samples (76%, 22/29) than the 2-LTR assay (41%, 12/29), (p = 0.0164). A trend towards lower uDNA levels was observed in aviremic vs viremic On-ART patients, reaching significance when we combined aviremic On-ART and HIC (controllers) vs Off-ART and viremic On-ART subjects (non-controllers) (p = 0.0003), whereas 2-LTR circle levels remained constant (p ≥ 0.2174). These data were supported by the high correlation found between uDNA and total DNA (r = 0.69, p < 0.001). CONCLUSIONS The great advantage of the U-assay is that, unlike the 2-LTR assay, it allows the accurate evaluation of the totality of uDNA that can still be measured even during successful ART when plasma viremia is below the cut-off of common clinical tests (< 50 copies/mL) and 2-LTR circles are more likely to be under the quantification limit. UDNA measurement in blood cells may be used as a biomarker to reveal a so far hidden or underestimated viral reservoir. The potential clinical relevance of uDNA quantification may lead to improvements in diagnostic methods to support clinical strategies.
Collapse
Affiliation(s)
- Chiara Orlandi
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Benedetta Canovari
- Malattie Infettive, Azienda Ospedaliera Ospedali Riuniti Marche Nord, Pesaro, Italy
| | | | - Francesco Marras
- Division of Infectious Diseases, Ospedale Policlinico S. Martino IRCCS, Genoa, Italy
| | - Zeno Pasquini
- Malattie Infettive, Azienda Ospedaliera Ospedali Riuniti Marche Nord, Pesaro, Italy
- Dipartimento di Scienze Biomediche e Sanità Pubblica, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco Barchiesi
- Malattie Infettive, Azienda Ospedaliera Ospedali Riuniti Marche Nord, Pesaro, Italy
- Dipartimento di Scienze Biomediche e Sanità Pubblica, Università Politecnica delle Marche, Ancona, Italy
| | - Andrea De Maria
- Division of Infectious Diseases, Ospedale Policlinico S. Martino IRCCS, Genoa, Italy
- Department of Health Sciences, DISSAL, University of Genova, Genoa, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Anna Casabianca
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy.
| |
Collapse
|
39
|
Grudlewska-Buda K, Skowron K, Gospodarek-Komkowska E. Comparison of the intensity of biofilm formation by Listeria monocytogenes using classical culture-based method and digital droplet PCR. AMB Express 2020; 10:75. [PMID: 32303851 PMCID: PMC7165217 DOI: 10.1186/s13568-020-01007-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 04/07/2020] [Indexed: 12/27/2022] Open
Abstract
Listeria monocytogenes is a Gram-positive bacterium, commonly found in food, water or sewage. This microorganism is capable of forming biofilm on different surfaces such as steel, glass, polypropylene etc. Recently an increase in cases of listeriosis has been noted, making L. monocytogenes the important health threat. Therefore, there is a need for rapid and sensitive detection of this pathogen. This study aimed to compare the number of L. monocytogenes cells recovered from the biofilm (prepared on steel and polypropylene) using the detection and amplification of the hlyA gene (droplet digital PCR, ddPCR) and the classical culture method. The research material consisted of 96 L. monocytogenes strains. A total of 58 isolates were obtained from clinical samples and 38 isolates derived from the municipal sewage treatment plant. Additionally, the reference strain ATCC®19111™ (WDCM00020) was used. The Pearson correlation coefficient for the results obtained by the classical culture-based method and ddPCR was 0.864 and 0.725, for biofilms produced on AISI 304 stainless steel surface and the polypropylene surface, respectively. Correlations were statistically significant (p ≤ 0.001), indicating that the ddPCR technique is an effective tool for the assessment of bacteria number in the biofilm.
Collapse
Affiliation(s)
- Katarzyna Grudlewska-Buda
- Department of Microbiology, Nicolaus Copernicus University in Toruń, Collegium Medicum of L. Rydygier in Bydgoszcz, Bydgoszcz, Poland
| | - Krzysztof Skowron
- Department of Microbiology, Nicolaus Copernicus University in Toruń, Collegium Medicum of L. Rydygier in Bydgoszcz, Bydgoszcz, Poland
| | - Eugenia Gospodarek-Komkowska
- Department of Microbiology, Nicolaus Copernicus University in Toruń, Collegium Medicum of L. Rydygier in Bydgoszcz, Bydgoszcz, Poland
| |
Collapse
|
40
|
Thomas J, Ruggiero A, Paxton WA, Pollakis G. Measuring the Success of HIV-1 Cure Strategies. Front Cell Infect Microbiol 2020; 10:134. [PMID: 32318356 PMCID: PMC7154081 DOI: 10.3389/fcimb.2020.00134] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/13/2020] [Indexed: 01/10/2023] Open
Abstract
HIV-1 eradication strategies aim to achieve viral remission in the absence of antiretroviral therapy (ART). The development of an HIV-1 cure remains challenging due to the latent reservoir (LR): long-lived CD4 T cells that harbor transcriptionally silent HIV-1 provirus. The LR is stable despite years of suppressive ART and is the source of rebound viremia following therapy interruption. Cure strategies such as "shock and kill" aim to eliminate or reduce the LR by reversing latency, exposing the infected cells to clearance via the immune response or the viral cytopathic effect. Alternative strategies include therapeutic vaccination, which aims to prime the immune response to facilitate control of the virus in the absence of ART. Despite promising advances, these strategies have been unable to significantly reduce the LR or increase the time to viral rebound but have provided invaluable insight in the field of HIV-1 eradication. The development and assessment of an HIV-1 cure requires robust assays that can measure the LR with sufficient sensitivity to detect changes that may occur following treatment. The viral outgrowth assay (VOA) is considered the gold standard method for LR quantification due to its ability to distinguish intact and defective provirus. However, the VOA is time consuming and resource intensive, therefore several alternative assays have been developed to bridge the gap between practicality and accuracy. Whilst a cure for HIV-1 infection remains elusive, recent advances in our understanding of the LR and methods for its eradication have offered renewed hope regarding achieving ART free viral remission.
Collapse
Affiliation(s)
- Jordan Thomas
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Alessandra Ruggiero
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Immune and Infectious Disease Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, Rome, Italy
| | - William A Paxton
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Georgios Pollakis
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
41
|
Shin W, Kim H, Oh DY, Kim DH, Han K. Quantitative evaluation of the molecular marker using droplet digital PCR. Genomics Inform 2020; 18:e4. [PMID: 32224837 PMCID: PMC7120350 DOI: 10.5808/gi.2020.18.1.e4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/05/2019] [Indexed: 11/24/2022] Open
Abstract
Transposable elements (TEs) constitute approximately half of Bovine genome. They can be a powerful species-specific marker without regression mutations by the structure variation (SV) at the time of genomic evolution. In a previous study, we identified the Hanwoo-specific SV that was generated by a TE–association deletion event using traditional PCR method and Sanger sequencing validation. It could be used as a molecular marker to distinguish different cattle breeds (i.e., Hanwoo vs. Holstein). However, PCR is defective with various final copy quantifications from every sample. Thus, we applied to the droplet digital PCR (ddPCR) platform for accurate quantitative detection of the Hanwoo-specific SV. Although samples have low allele frequency variation within Hanwoo population, ddPCR could perform high sensitive detection with absolute quantification. We aimed to use ddPCR for more accurate quantification than PCR. We suggest that the ddPCR platform is applicable for the quantitative evaluation of molecular markers.
Collapse
Affiliation(s)
- Wonseok Shin
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea.,Center for Bio-Medical Engineering Core Facility, Dankook University, Cheonan 31116, Korea
| | - Haneul Kim
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea.,Center for Bio-Medical Engineering Core Facility, Dankook University, Cheonan 31116, Korea
| | - Dong-Yep Oh
- Livestock Research Institute, Yeongju 36052, Korea
| | - Dong Hee Kim
- Department of Anesthesiology and Pain Management, Dankook University College of Medicine, Cheonan 31116, Korea
| | - Kyudong Han
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea.,Center for Bio-Medical Engineering Core Facility, Dankook University, Cheonan 31116, Korea
| |
Collapse
|
42
|
Fidler S, Stöhr W, Pace M, Dorrell L, Lever A, Pett S, Kinloch-de Loes S, Fox J, Clarke A, Nelson M, Thornhill J, Khan M, Fun A, Bandara M, Kelly D, Kopycinski J, Hanke T, Yang H, Bennett R, Johnson M, Howell B, Barnard R, Wu G, Kaye S, Wills M, Babiker A, Frater J. Antiretroviral therapy alone versus antiretroviral therapy with a kick and kill approach, on measures of the HIV reservoir in participants with recent HIV infection (the RIVER trial): a phase 2, randomised trial. Lancet 2020; 395:888-898. [PMID: 32085823 DOI: 10.1016/s0140-6736(19)32990-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/25/2019] [Accepted: 11/15/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Antiretroviral therapy (ART) cannot cure HIV infection because of a persistent reservoir of latently infected cells. Approaches that force HIV transcription from these cells, making them susceptible to killing-termed kick and kill regimens-have been explored as a strategy towards an HIV cure. RIVER is the first randomised trial to determine the effect of ART-only versus ART plus kick and kill on markers of the HIV reservoir. METHODS This phase 2, open-label, multicentre, randomised, controlled trial was undertaken at six clinical sites in the UK. Patients aged 18-60 years who were confirmed as HIV-positive within a maximum of the past 6 months and started ART within 1 month from confirmed diagnosis were randomly assigned by a computer generated randomisation list to receive ART-only (control) or ART plus the histone deacetylase inhibitor vorinostat (the kick) and replication-deficient viral vector T-cell inducing vaccines encoding conserved HIV sequences ChAdV63. HIVconsv-prime and MVA.HIVconsv-boost (the kill; ART + V + V; intervention). The primary endpoint was total HIV DNA isolated from peripheral blood CD4+ T-cells at weeks 16 and 18 after randomisation. Analysis was by intention to treat. This trial is registered with ClinicalTrials.gov, NCT02336074. FINDINGS Between June 14, 2015 and Jul 11, 2017, 60 men with HIV were randomly assigned to receive either an ART-only (n=30) or an ART + V + V (n=30) regimen; all 60 participants completed the study, with no loss-to-follow-up. Mean total HIV DNA at weeks 16 and 18 after randomisation was 3·02 log10 copies HIV DNA per 106 CD4+ T-cells in the ART-only group versus 3·06 log10 copies HIV DNA per 106 CD4+ T-cells in ART + V + V group, with no statistically significant difference between the two groups (mean difference of 0·04 log10 copies HIV DNA per 106 CD4+ T-cells [95% CI -0·03 to 0·11; p=0·26]). There were no intervention-related serious adverse events. INTERPRETATION This kick and kill approach conferred no significant benefit compared with ART alone on measures of the HIV reservoir. Although this does not disprove the efficacy kick and kill strategy, for future trials enhancement of both kick and kill agents will be required. FUNDING Medical Research Council (MR/L00528X/1).
Collapse
Affiliation(s)
- Sarah Fidler
- Department of Infectious Disease, Imperial College London, London, UK; NIHR Imperial Biomedical Research Centre, London, UK.
| | - Wolfgang Stöhr
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | - Matt Pace
- Nuffield Department of Medicine, Oxford University, UK; Nuffield Department of Medicine Oxford Martin School, Oxford, UK
| | - Lucy Dorrell
- Nuffield Department of Medicine, Oxford University, UK; Nuffield Department of Medicine, Oxford NIHR Biomedical Research Centre, Oxford, UK
| | - Andrew Lever
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sarah Pett
- Medical Research Council Clinical Trials Unit, University College London, London, UK; Institute for Global Health, University College London, London, UK; Mortimer Market Centre, London, UK
| | - Sabine Kinloch-de Loes
- Department of Infection and Immunity, Royal Free Hospital, London, UK; University College London, London, UK
| | - Julie Fox
- Department of Genitourinary Medicine and Infectious Disease, Guys and St Thomas' NHS Trust, London, UK; Department of Genitourinary Medicine and Infectious Disease, NIHR Biomedical Research Centre, Kings College London, London, UK
| | - Amanda Clarke
- Elton John Centre, Brighton, UK; Department of HIV and Sexual Health, Sussex University Hospital, Brighton, UK; Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Mark Nelson
- Chelsea and Westminster Hospital, Department of HIV Medicine, Imperial College London London, UK
| | - John Thornhill
- Department of Infectious Disease, Imperial College London, London, UK; NIHR Imperial Biomedical Research Centre, London, UK
| | - Maryam Khan
- Department of Infectious Disease, Imperial College London, London, UK; NIHR Imperial Biomedical Research Centre, London, UK
| | - Axel Fun
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | | | - Jakub Kopycinski
- Nuffield Department of Medicine, Oxford University, UK; Nuffield Department of Medicine, Oxford NIHR Biomedical Research Centre, Oxford, UK
| | - Tomáš Hanke
- Nuffield Department of Medicine, Oxford University, UK; International Research Center for Medical Sciences, Kumamoto University, Japan
| | - Hongbing Yang
- Nuffield Department of Medicine, Oxford University, UK; Nuffield Department of Medicine, Oxford NIHR Biomedical Research Centre, Oxford, UK
| | - Rachel Bennett
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | | | - Bonnie Howell
- Department of Infectious Disease and Vaccines, Merck and Co, West Point, PA, USA
| | - Richard Barnard
- Global Regulatory Affairs and Clinical Safety, Merck and Co, North Wales, PA, USA
| | - Guoxin Wu
- Department of Infectious Disease and Vaccines, Merck and Co, West Point, PA, USA
| | - Steve Kaye
- Department of Infectious Disease, Imperial College London, London, UK
| | - Mark Wills
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Abdel Babiker
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | - John Frater
- Nuffield Department of Medicine, Oxford University, UK; Nuffield Department of Medicine, Oxford NIHR Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
43
|
Kint S, Trypsteen W, De Spiegelaere W, Malatinkova E, Kinloch-de Loes S, De Meyer T, Van Criekinge W, Vandekerckhove L. Underestimated effect of intragenic HIV-1 DNA methylation on viral transcription in infected individuals. Clin Epigenetics 2020; 12:36. [PMID: 32111236 PMCID: PMC7049218 DOI: 10.1186/s13148-020-00829-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/16/2020] [Indexed: 12/13/2022] Open
Abstract
Background The HIV-1 proviral genome harbors multiple CpG islands (CpGIs), both in the promoter and intragenic regions. DNA methylation in the promoter region has been shown to be heavily involved in HIV-1 latency regulation in cultured cells. However, its exact role in proviral transcriptional regulation in infected individuals is poorly understood or characterized. Moreover, methylation at intragenic CpGIs has never been studied in depth. Results A large, well-characterized HIV-1 patient cohort (n = 72), consisting of 17 long-term non-progressors and 8 recent seroconverters (SRCV) without combination antiretroviral therapy (cART), 15 early cART-treated, and 32 late cART-treated patients, was analyzed using a next-generation bisulfite sequencing DNA methylation method. In general, we observed low level of promoter methylation and higher levels of intragenic methylation. Additionally, SRCV showed increased promoter methylation and decreased intragenic methylation compared with the other patient groups. This data indicates that increased intragenic methylation could be involved in proviral transcriptional regulation. Conclusions Contrasting in vitro studies, our results indicate that intragenic hypermethylation of HIV-1 proviral DNA is an underestimated factor in viral control in HIV-1-infected individuals, showing the importance of analyzing the complete proviral genome in future DNA methylation studies.
Collapse
Affiliation(s)
- Sam Kint
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, Medical Research Building 2, 9000, Ghent, Belgium.,Biobix, Department of Data Analysis and Mathematical Modelling, Faculty of Bio-science Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Wim Trypsteen
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, Medical Research Building 2, 9000, Ghent, Belgium
| | - Ward De Spiegelaere
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Eva Malatinkova
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, Medical Research Building 2, 9000, Ghent, Belgium
| | - Sabine Kinloch-de Loes
- Division of Infection and Immunity, Royal Free Hospital, Royal Free Campus, University College London, Pont St, Hampstead, London, NW3 2QG, UK
| | - Tim De Meyer
- Biobix, Department of Data Analysis and Mathematical Modelling, Faculty of Bio-science Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Wim Van Criekinge
- Biobix, Department of Data Analysis and Mathematical Modelling, Faculty of Bio-science Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, Medical Research Building 2, 9000, Ghent, Belgium.
| |
Collapse
|
44
|
Jahne MA, Brinkman NE, Keely SP, Zimmerman BD, Wheaton EA, Garland JL. Droplet digital PCR quantification of norovirus and adenovirus in decentralized wastewater and graywater collections: Implications for onsite reuse. WATER RESEARCH 2020; 169:115213. [PMID: 31671297 PMCID: PMC7017454 DOI: 10.1016/j.watres.2019.115213] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/08/2019] [Accepted: 10/17/2019] [Indexed: 05/04/2023]
Abstract
Risk-based treatment of onsite wastewaters for decentralized reuse requires information on the occurrence and density of pathogens in source waters, which differ from municipal wastewater due to scaling and dilution effects in addition to variable source contributions. In this first quantitative report of viral enteric pathogens in onsite-collected graywater and wastewater, untreated graywater (n = 50 samples) and combined wastewater (i.e., including blackwater; n = 28) from three decentralized collection systems were analyzed for two norovirus genogroups (GI/GII) and human adenoviruses using droplet digital polymerase chain reaction (ddPCR). Compared to traditional quantitative PCR (qPCR), which had insufficient sensitivity to quantify viruses in graywater, ddPCR allowed quantification of norovirus GII and adenovirus in 4% and 14% of graywater samples, respectively (none quantifiable for norovirus GI). Norovirus GII was routinely quantifiable in combined wastewater by either PCR method (96% of samples), with well-correlated results between the analyses (R2 = 0.96) indicating a density range of 5.2-7.9 log10 genome copies/L. These concentrations are greater than typically reported in centralized municipal wastewater, yet agree well with an epidemiology-based model previously used to develop pathogen log-reduction targets (LRTs) for decentralized non-potable water systems. Results emphasize the unique quality of onsite wastewaters, supporting the previous LRTs and further quantitative microbial risk assessment (QMRA) of decentralized water reuse.
Collapse
Affiliation(s)
- Michael A Jahne
- Office of Research and Development, U.S. Environmental Protection Agency, 26 W. Martin Luther King Dr., Cincinnati, OH, 45268, USA.
| | - Nichole E Brinkman
- Office of Research and Development, U.S. Environmental Protection Agency, 26 W. Martin Luther King Dr., Cincinnati, OH, 45268, USA
| | - Scott P Keely
- Office of Research and Development, U.S. Environmental Protection Agency, 26 W. Martin Luther King Dr., Cincinnati, OH, 45268, USA
| | - Brian D Zimmerman
- Office of Research and Development, U.S. Environmental Protection Agency, 26 W. Martin Luther King Dr., Cincinnati, OH, 45268, USA
| | - Emily A Wheaton
- Office of Research and Development, U.S. Environmental Protection Agency, 26 W. Martin Luther King Dr., Cincinnati, OH, 45268, USA
| | - Jay L Garland
- Office of Research and Development, U.S. Environmental Protection Agency, 26 W. Martin Luther King Dr., Cincinnati, OH, 45268, USA
| |
Collapse
|
45
|
Anderson EM, Simonetti FR, Gorelick RJ, Hill S, Gouzoulis MA, Bell J, Rehm C, Pérez L, Boritz E, Wu X, Wells D, Hughes SH, Rao V, Coffin JM, Kearney MF, Maldarelli F. Dynamic Shifts in the HIV Proviral Landscape During Long Term Combination Antiretroviral Therapy: Implications for Persistence and Control of HIV Infections. Viruses 2020; 12:v12020136. [PMID: 31991737 PMCID: PMC7077288 DOI: 10.3390/v12020136] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/13/2020] [Accepted: 01/16/2020] [Indexed: 12/19/2022] Open
Abstract
Combination antiretroviral therapy (cART) controls but does not eradicate HIV infection; HIV persistence is the principal obstacle to curing infections. The proportion of defective proviruses increases during cART, but the dynamics of this process are not well understood, and a quantitative analysis of how the proviral landscape is reshaped after cART is initiated is critical to understanding how HIV persists. Here, we studied longitudinal samples from HIV infected individuals undergoing long term cART using multiplexed Droplet Digital PCR (ddPCR) approaches to quantify the proportion of deleted proviruses in lymphocytes. In most individuals undergoing cART, HIV proviruses that contain gag are lost more quickly than those that lack gag. Increases in the fraction of gag-deleted proviruses occurred only after 1–2 years of therapy, suggesting that the immune system, and/or toxicity of viral re-activation helps to gradually shape the proviral landscape. After 10–15 years on therapy, there were as many as 3.5–5 times more proviruses in which gag was deleted or highly defective than those containing intact gag. We developed a provirus-specific ddPCR approach to quantify individual clones. Investigation of a clone of cells containing a deleted HIV provirus integrated in the HORMAD2 gene revealed that the cells underwent a massive expansion shortly after cART was initiated until the clone, which was primarily in effector memory cells, dominated the population of proviruses for over 6 years. The expansion of this HIV-infected clone had substantial effects on the overall proviral population.
Collapse
Affiliation(s)
- Elizabeth M. Anderson
- HIV Dynamics and Replication Program, NCI, NIH, Frederick, MD 21702, USA; (E.M.A.); (F.R.S.); (S.H.); (M.A.G.); (S.H.H.); (M.F.K.)
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA;
| | - Francesco R. Simonetti
- HIV Dynamics and Replication Program, NCI, NIH, Frederick, MD 21702, USA; (E.M.A.); (F.R.S.); (S.H.); (M.A.G.); (S.H.H.); (M.F.K.)
| | - Robert J. Gorelick
- Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (R.J.G.); (J.B.)
| | - Shawn Hill
- HIV Dynamics and Replication Program, NCI, NIH, Frederick, MD 21702, USA; (E.M.A.); (F.R.S.); (S.H.); (M.A.G.); (S.H.H.); (M.F.K.)
| | - Monica A. Gouzoulis
- HIV Dynamics and Replication Program, NCI, NIH, Frederick, MD 21702, USA; (E.M.A.); (F.R.S.); (S.H.); (M.A.G.); (S.H.H.); (M.F.K.)
| | - Jennifer Bell
- Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (R.J.G.); (J.B.)
| | - Catherine Rehm
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD 20814, USA;
| | - Liliana Pérez
- Virus Persistence and Dynamics Section, VRC, NIAID, NIH, Bethesda, MD 20814, USA; (L.P.); (E.B.)
| | - Eli Boritz
- Virus Persistence and Dynamics Section, VRC, NIAID, NIH, Bethesda, MD 20814, USA; (L.P.); (E.B.)
| | - Xiaolin Wu
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (X.W.); (D.W.)
| | - Daria Wells
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (X.W.); (D.W.)
| | - Stephen H. Hughes
- HIV Dynamics and Replication Program, NCI, NIH, Frederick, MD 21702, USA; (E.M.A.); (F.R.S.); (S.H.); (M.A.G.); (S.H.H.); (M.F.K.)
| | - Venigalla Rao
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA;
| | - John M. Coffin
- Department of Biology, Tufts University, Boston, MA 02155, USA;
| | - Mary F. Kearney
- HIV Dynamics and Replication Program, NCI, NIH, Frederick, MD 21702, USA; (E.M.A.); (F.R.S.); (S.H.); (M.A.G.); (S.H.H.); (M.F.K.)
| | - Frank Maldarelli
- HIV Dynamics and Replication Program, NCI, NIH, Frederick, MD 21702, USA; (E.M.A.); (F.R.S.); (S.H.); (M.A.G.); (S.H.H.); (M.F.K.)
- Correspondence: ; Tel.: +01-301-846-5611
| |
Collapse
|
46
|
Salipante SJ, Jerome KR. Digital PCR—An Emerging Technology with Broad Applications in Microbiology. Clin Chem 2019; 66:117-123. [DOI: 10.1373/clinchem.2019.304048] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/18/2019] [Indexed: 01/10/2023]
Abstract
Abstract
BACKGROUND
The PCR and its variant, quantitative PCR (qPCR), have revolutionized the practice of clinical microbiology. Continued advancements in PCR have led to a new derivative, digital PCR (dPCR), which promises to address certain limitations inherent to qPCR.
CONTENT
Here we highlight the important technical differences between qPCR and dPCR, and the potential advantages and disadvantages of each. We then review specific situations in which dPCR has been implemented in clinical microbiology and the results of such applications. Finally, we attempt to place dPCR in the context of other emerging technologies relevant to the clinical laboratory, including next-generation sequencing.
SUMMARY
dPCR offers certain clear advantages over traditional qPCR, but these are to some degree offset by limitations of the technology, at least as currently practiced. Laboratories considering implementation of dPCR should carefully weigh the potential advantages and disadvantages of this powerful technique for each specific application planned.
Collapse
Affiliation(s)
| | - Keith R Jerome
- Department of Laboratory Medicine, University of Washington, Seattle, WA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
47
|
Falcinelli SD, Ceriani C, Margolis DM, Archin NM. New Frontiers in Measuring and Characterizing the HIV Reservoir. Front Microbiol 2019; 10:2878. [PMID: 31921056 PMCID: PMC6930150 DOI: 10.3389/fmicb.2019.02878] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/28/2019] [Indexed: 12/13/2022] Open
Abstract
A cure for HIV infection remains elusive due to the persistence of replication-competent HIV proviral DNA during suppressive antiretroviral therapy (ART). With the exception of rare elite or post-treatment controllers of viremia, withdrawal of ART invariably results in the rebound of viremia and progression of HIV disease. A thorough understanding of the reservoir is necessary to develop new strategies in order to reduce or eliminate the reservoir. However, there is significant heterogeneity in the sequence composition, genomic location, stability, and expression of the HIV reservoir both within and across individuals, and a majority of proviral sequences are replication-defective. These factors, and the low frequency of persistently infected cells in individuals on suppressive ART, make understanding the reservoir and its response to experimental reservoir reduction interventions challenging. Here, we review the characteristics of the HIV reservoir, state-of-the-art assays to measure and characterize the reservoir, and how these assays can be applied to accurately detect reductions in reservoir during efforts to develop a cure for HIV infection. In particular, we highlight recent advances in the development of direct measures of provirus, including intact proviral DNA assays and full-length HIV DNA sequencing with integration site analysis. We also focus on novel techniques to quantitate persistent and inducible HIV, including RNA sequencing and RNA/gag protein staining techniques, as well as modified viral outgrowth methods that seek to improve upon throughput, sensitivity and dynamic range.
Collapse
Affiliation(s)
- Shane D Falcinelli
- UNC HIV Cure Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Cristina Ceriani
- UNC HIV Cure Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - David M Margolis
- UNC HIV Cure Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nancie M Archin
- UNC HIV Cure Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
48
|
Jacobs JL, Halvas EK, Tosiano MA, Mellors JW. Persistent HIV-1 Viremia on Antiretroviral Therapy: Measurement and Mechanisms. Front Microbiol 2019; 10:2383. [PMID: 31681237 PMCID: PMC6804636 DOI: 10.3389/fmicb.2019.02383] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 10/01/2019] [Indexed: 12/28/2022] Open
Abstract
HIV-1 viremia persists at low-levels despite clinically effective antiretroviral therapy (ART). Here we review new methods to quantify and characterize persistent viremia at the single genome level, and discuss the mechanisms of persistence including clonal expansion of infected cells and tissue origins of viremia. A deeper understanding of how viremia persists on ART is critically important to the design of therapies to eliminate viremia and achieve a functional cure for HIV-1.
Collapse
Affiliation(s)
- Jana L Jacobs
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Elias K Halvas
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Melissa A Tosiano
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - John W Mellors
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
49
|
O'Hara R, Tedone E, Ludlow A, Huang E, Arosio B, Mari D, Shay JW. Quantitative mitochondrial DNA copy number determination using droplet digital PCR with single-cell resolution. Genome Res 2019; 29:1878-1888. [PMID: 31548359 PMCID: PMC6836731 DOI: 10.1101/gr.250480.119] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 09/20/2019] [Indexed: 12/16/2022]
Abstract
Mitochondria are involved in a number of diverse cellular functions, including energy production, metabolic regulation, apoptosis, calcium homeostasis, cell proliferation, and motility, as well as free radical generation. Mitochondrial DNA (mtDNA) is present at hundreds to thousands of copies per cell in a tissue-specific manner. mtDNA copy number also varies during aging and disease progression and therefore might be considered as a biomarker that mirrors alterations within the human body. Here, we present a new quantitative, highly sensitive droplet digital PCR (ddPCR) method, droplet digital mitochondrial DNA measurement (ddMDM), to measure mtDNA copy number not only from cell populations but also from single cells. Our developed assay can generate data in as little as 3 h, is optimized for 96-well plates, and also allows the direct use of cell lysates without the need for DNA purification or nuclear reference genes. We show that ddMDM is able to detect differences between samples whose mtDNA copy number was close enough as to be indistinguishable by other commonly used mtDNA quantitation methods. By utilizing ddMDM, we show quantitative changes in mtDNA content per cell across a wide variety of physiological contexts including cancer progression, cell cycle progression, human T cell activation, and human aging.
Collapse
Affiliation(s)
- Ryan O'Hara
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Enzo Tedone
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Andrew Ludlow
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ejun Huang
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Beatrice Arosio
- Geriatric Unit, Department of Medical Sciences and Community Health, University of Milan, 20122 Milan, Italy.,Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Daniela Mari
- Geriatric Unit, Department of Medical Sciences and Community Health, University of Milan, 20122 Milan, Italy.,Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Jerry W Shay
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
50
|
Del Giudice I, Raponi S, Della Starza I, De Propris MS, Cavalli M, De Novi LA, Cappelli LV, Ilari C, Cafforio L, Guarini A, Foà R. Minimal Residual Disease in Chronic Lymphocytic Leukemia: A New Goal? Front Oncol 2019; 9:689. [PMID: 31555576 PMCID: PMC6727319 DOI: 10.3389/fonc.2019.00689] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022] Open
Abstract
In chronic lymphocytic leukemia (CLL), there is a growing interest for minimal residual disease (MRD) monitoring, due to the availability of drug combinations capable of unprecedented complete clinical responses. The standardized and most commonly applied methods to assess MRD in CLL are based on flow cytometry (FCM) and, to a lesser extent, real-time quantitative PCR (RQ-PCR) with allele-specific oligonucleotide (ASO) primers of immunoglobulin heavy chain genes (IgH). Promising results are being obtained using droplet digital PCR (ddPCR) and next generation sequencing (NGS)-based approaches, with some advantages and a potential higher sensitivity compared to the standardized methodologies. Plasma cell-free DNA can also be explored as a more precise measure of residual disease from all different compartments, including the lymph nodes. From a clinical point of view, CLL MRD quantification has proven an independent prognostic marker of progression-free survival (PFS) and overall survival (OS) after chemoimmunotherapy as well as after allogeneic transplantation. In the era of mechanism-driven drugs, the paradigms of CLL treatment are being revolutionized, challenging the use of chemoimmunotherapy even in first-line. The continuous administration of ibrutinib single agent has led to prolonged PFS and OS in relapsed/refractory and treatment naïve CLL, including those with TP53 deletion/mutation or unmutated IGHV genes, though the clinical responses are rarely complete. More recently, chemo-free combinations of venetoclax+rituximab, venetoclax+obinutuzumab or ibrutinib+venetoclax have been shown capable of inducing undetectable MRD in the bone marrow, opening the way to protocols exploring a MRD-based duration of treatment, aiming at disease eradication. Thus, beside a durable disease control desirable particularly for older patients and/or for those with comorbidities, a MRD-negative complete remission is becoming a realistic prospect for CLL patients in an attempt to obtain a long-lasting eradication and possibly cure of the disease. Here we discuss the standardized and innovative technical approaches for MRD detection in CLL, the clinical impact of MRD monitoring in chemoimmunotherapy and chemo-free trials and the future clinical implications of MRD monitoring in CLL patients outside of clinical trials.
Collapse
Affiliation(s)
- Ilaria Del Giudice
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Sara Raponi
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.,GIMEMA Foundation, Rome, Italy
| | - Maria Stefania De Propris
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Marzia Cavalli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia Anna De Novi
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Luca Vincenzo Cappelli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Caterina Ilari
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Luciana Cafforio
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Anna Guarini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|