Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Int J Mol Sci ; 24(9)2023 May 03.
Article in English | MEDLINE | ID: covidwho-2315346

ABSTRACT

Severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) may impair immune modulating host microRNAs, causing severe disease. Our objectives were to determine the salivary miRNA profile in children with SARS-CoV-2 infection at presentation and compare the expression in those with and without severe outcomes. Children <18 years with SARS-CoV-2 infection evaluated at two hospitals between March 2021 and February 2022 were prospectively enrolled. Severe outcomes included respiratory failure, shock or death. Saliva microRNAs were quantified with RNA sequencing. Data on 197 infected children (severe = 45) were analyzed. Of the known human miRNAs, 1606 (60%) were measured and compared across saliva samples. There were 43 miRNAs with ≥2-fold difference between severe and non-severe cases (adjusted p-value < 0.05). The majority (31/43) were downregulated in severe cases. The largest between-group differences involved miR-4495, miR-296-5p, miR-548ao-3p and miR-1273c. These microRNAs displayed enrichment for 32 gene ontology pathways including viral processing and transforming growth factor beta and Fc-gamma receptor signaling. In conclusion, salivary miRNA levels are perturbed in children with severe COVID-19, with the majority of miRNAs being down regulated. Further studies are required to validate and determine the utility of salivary miRNAs as biomarkers of severe COVID-19.


Subject(s)
COVID-19 , MicroRNAs , Humans , Child , Saliva/metabolism , COVID-19/genetics , COVID-19/metabolism , SARS-CoV-2/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction
2.
Int J Mol Sci ; 24(6)2023 Mar 09.
Article in English | MEDLINE | ID: covidwho-2281808

ABSTRACT

Saliva is gaining increasing attention as a source of biomarkers due to non-invasive and undemanding collection access. Extracellular vesicles (EVs) are nano-sized, cell-released particles that contain molecular information about their parent cells. In this study, we developed methods for saliva biomarker candidate identification using EV-isolation and proteomic evaluation. We used pooled saliva samples for assay development. EVs were isolated using membrane affinity-based methods followed by their characterization using nanoparticle tracking analysis and transmission electron microscopy. Subsequently, both saliva and saliva-EVs were successfully analyzed using proximity extension assay and label-free quantitative proteomics. Saliva-EVs had a higher purity than plasma-EVs, based on the expression of EV-proteins and albumin. The developed methods could be used for the analysis of individual saliva samples from amyotrophic lateral sclerosis (ALS) patients and controls (n = 10 each). The starting volume ranged from 2.1 to 4.9 mL and the amount of total isolated EV-proteins ranged from 5.1 to 42.6 µg. Although no proteins were significantly differentially expressed between the two groups, there was a trend for a downregulation of ZNF428 in ALS-saliva-EVs and an upregulation of IGLL1 in ALS saliva. In conclusion, we have developed a robust workflow for saliva and saliva-EV analysis and demonstrated its technical feasibility for biomarker discovery.


Subject(s)
Amyotrophic Lateral Sclerosis , Extracellular Vesicles , Humans , Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/metabolism , Pilot Projects , Proteomics/methods , Saliva/metabolism , Extracellular Vesicles/metabolism , Biomarkers/metabolism
3.
Metabolomics ; 18(11): 81, 2022 Oct 22.
Article in English | MEDLINE | ID: covidwho-2085518

ABSTRACT

INTRODUCTION: Coronavirus disease 2019 (COVID-19) is strongly linked to dysregulation of various molecular, cellular, and physiological processes that change abundance of different biomolecules including metabolites that may be ultimately used as biomarkers for disease progression and severity. It is important at early stage to readily distinguish those patients that are likely to progress to moderate and severe stages. OBJECTIVES: This study aimed to investigate the utility of saliva and plasma metabolomic profiles as a potential parameter for risk stratifying COVID-19 patients. METHOD: LC-MS/MS-based untargeted metabolomics were used to profile the changes in saliva and plasma metabolomic profiles of COVID-19 patients with different severities. RESULTS: Saliva and plasma metabolites were screened in 62 COVID-19 patients and 18 non-infected controls. The COVID-19 group included 16 severe, 15 moderate, 16 mild, and 15 asymptomatic cases. Thirty-six differential metabolites were detected in COVID-19 versus control comparisons. SARS-CoV-2 induced metabolic derangement differed with infection severity. The metabolic changes were identified in saliva and plasma, however, saliva showed higher intensity of metabolic changes. Levels of saliva metabolites such as sphingosine and kynurenine were significantly different between COVID-19 infected and non-infected individuals; while linoleic acid and Alpha-ketoisovaleric acid were specifically increased in severe compared to non-severe patients. As expected, the two prognostic biomarkers of C-reactive protein and D-dimer were negatively correlated with sphingosine and 5-Aminolevulinic acid, and positively correlated with L-Tryptophan and L-Kynurenine. CONCLUSION: Saliva disease-specific and severity-specific metabolite could be employed as potential COVID-19 diagnostic and prognostic biomarkers.


Subject(s)
COVID-19 , Humans , Metabolomics , SARS-CoV-2 , Saliva/metabolism , Chromatography, Liquid , Kynurenine/metabolism , Tryptophan/metabolism , C-Reactive Protein/metabolism , Sphingosine , Linoleic Acid/metabolism , Aminolevulinic Acid/metabolism , Tandem Mass Spectrometry , Severity of Illness Index , Biomarkers
4.
Clin Chim Acta ; 537: 26-37, 2022 Dec 01.
Article in English | MEDLINE | ID: covidwho-2060488

ABSTRACT

BACKGROUND AND AIM: SARS-CoV-2 infection spawns from an asymptomatic condition to a fatal disease. Age, comorbidities, and several blood biomarkers are associated with infection outcome. We searched for biomarkers by untargeted and targeted proteomic analysis of saliva, a source of viral particles and host proteins. METHODS: Saliva samples from 19 asymptomatic and 16 symptomatic SARS-CoV-2 infected subjects, and 20 controls were analyzed by LC-MS/MS for untargeted peptidomic (flow through of 10 kDa filter) and proteomic (trypsin digestion of filter retained proteins) profiling. RESULTS: Peptides from 53 salivary proteins were identified. ADF was detected only in controls, while IL1RA only in infected subjects. PRPs, DSC2, FABP5, his-1, IL1RA, PRH1, STATH, SMR3B, ANXA1, MUC7, ACTN4, IGKV1-33 and TGM3 were significantly different between asymptomatic and symptomatic subjects. Retained proteins were 117, being 11 highly different between asymptomatic and symptomatic (fold change ≥2 or ≤-2). After validation by LC-MS/MS-SRM (selected reaction monitoring analysis), the most significant discriminant proteins at PCA were IL1RA, CYSTB, S100A8, S100A9, CA6, and FABP5. CONCLUSIONS: The differentially abundant proteins involved in innate immunity (S100 proteins), taste (CA6 and cystatins), and viral binding to the host (FABP5), appear to be of interest for use as potential biomarkers and drugs targets.


Subject(s)
COVID-19 , Proteomics , Humans , Chromatography, Liquid , Taste Perception , SARS-CoV-2 , Taste , Tandem Mass Spectrometry , Saliva/metabolism , Biomarkers/metabolism , Immunity, Innate , Fatty Acid-Binding Proteins/metabolism , Transglutaminases/metabolism
5.
PLoS One ; 17(9): e0274967, 2022.
Article in English | MEDLINE | ID: covidwho-2039439

ABSTRACT

BACKGROUND: The COVID-19 pandemic is likely to represent an ongoing global health issue given the potential for new variants, vaccine escape and the low likelihood of eliminating all reservoirs of the disease. Whilst diagnostic testing has progressed at a fast pace, the metabolic drivers of outcomes-and whether markers can be found in different biofluids-are not well understood. Recent research has shown that serum metabolomics has potential for prognosis of disease progression. In a hospital setting, collection of saliva samples is more convenient for both staff and patients, and therefore offers an alternative sampling matrix to serum. METHODS: Saliva samples were collected from hospitalised patients with clinical suspicion of COVID-19, alongside clinical metadata. COVID-19 diagnosis was confirmed using RT-PCR testing, and COVID-19 severity was classified using clinical descriptors (respiratory rate, peripheral oxygen saturation score and C-reactive protein levels). Metabolites were extracted and analysed using high resolution liquid chromatography-mass spectrometry, and the resulting peak area matrix was analysed using multivariate techniques. RESULTS: Positive percent agreement of 1.00 between a partial least squares-discriminant analysis metabolomics model employing a panel of 6 features (5 of which were amino acids, one that could be identified by formula only) and the clinical diagnosis of COVID-19 severity was achieved. The negative percent agreement with the clinical severity diagnosis was also 1.00, leading to an area under receiver operating characteristics curve of 1.00 for the panel of features identified. CONCLUSIONS: In this exploratory work, we found that saliva metabolomics and in particular amino acids can be capable of separating high severity COVID-19 patients from low severity COVID-19 patients. This expands the atlas of COVID-19 metabolic dysregulation and could in future offer the basis of a quick and non-invasive means of sampling patients, intended to supplement existing clinical tests, with the goal of offering timely treatment to patients with potentially poor outcomes.


Subject(s)
COVID-19 , Amino Acids/metabolism , Biomarkers/metabolism , C-Reactive Protein/metabolism , COVID-19/diagnosis , COVID-19 Testing , Chromatography, Liquid/methods , Humans , Mass Spectrometry/methods , Metabolomics/methods , Pandemics , Saliva/metabolism
6.
ACS Sens ; 7(10): 3006-3013, 2022 Oct 28.
Article in English | MEDLINE | ID: covidwho-2036753

ABSTRACT

Rapid and accurate identification of a pathogen is crucial for disease control and prevention of the epidemic of emerging infectious like SARS-CoV-2. However, no foolproof gold standard assay exists to date. Nucleic acid-based molecular diagnostic tests have been established for identifying COVID-19. However, viral RNAs are highly unstable in handling with poor laboratory procedures, leading to a false negative that accelerates the spread of the disease. Detection of the spike protein (S1) of the SARS-CoV-2 virus through a proper receptor, commonly used in antigen-based rapid testing kits, also suffers from false-negative predictions due to decreasing viral titers in clinical specimens. Organic field-effect transistor (OFET)-based sensors can be highly sensitive upon properly integrating receptors in the conducting channel. This work demonstrates how angiotensin-converting enzyme 2 (ACE2) molecules can be used as receptor molecules of the SARS-CoV-2 virus in the OFET platform. Integration of ACE2 molecules into pentacene grain boundaries has been studied through the statistical analysis of rough surfaces in terms of lateral correlation length and interface width. The uniform coating of ACE2 molecules has been confirmed through growth studies to achieve better ingress of the receptors into the conducting channel at the semiconductor/dielectric interface of OFETs. We have observed less than a minute detection time with 94% sensitivity, which is the highest reported value. The sensor works with a saliva sample, requiring no sample preparation or virus transfer medium. A prototype module developed for remote monitoring confirms the suitability for point-of-care (POC) application at large-scale testing in more crowded areas like airports, railway stations, shopping malls, etc.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Humans , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , COVID-19/diagnosis , Saliva/metabolism , Peptidyl-Dipeptidase A/metabolism
7.
Sci Rep ; 12(1): 11867, 2022 07 13.
Article in English | MEDLINE | ID: covidwho-1931494

ABSTRACT

The majority of metabolomics studies to date have utilised blood serum or plasma, biofluids that do not necessarily address the full range of patient pathologies. Here, correlations between serum metabolites, salivary metabolites and sebum lipids are studied for the first time. 83 COVID-19 positive and negative hospitalised participants provided blood serum alongside saliva and sebum samples for analysis by liquid chromatography mass spectrometry. Widespread alterations to serum-sebum lipid relationships were observed in COVID-19 positive participants versus negative controls. There was also a marked correlation between sebum lipids and the immunostimulatory hormone dehydroepiandrosterone sulphate in the COVID-19 positive cohort. The biofluids analysed herein were also compared in terms of their ability to differentiate COVID-19 positive participants from controls; serum performed best by multivariate analysis (sensitivity and specificity of 0.97), with the dominant changes in triglyceride and bile acid levels, concordant with other studies identifying dyslipidemia as a hallmark of COVID-19 infection. Sebum performed well (sensitivity 0.92; specificity 0.84), with saliva performing worst (sensitivity 0.78; specificity 0.83). These findings show that alterations to skin lipid profiles coincide with dyslipidaemia in serum. The work also signposts the potential for integrated biofluid analyses to provide insight into the whole-body atlas of pathophysiological conditions.


Subject(s)
COVID-19 , Sebum , Humans , Lipids/analysis , Metabolomics , Saliva/metabolism , Sebum/metabolism , Serum/chemistry
8.
Sci Rep ; 12(1): 3854, 2022 03 09.
Article in English | MEDLINE | ID: covidwho-1799575

ABSTRACT

The outbreak of COVID-19 caused by infection with SARS-CoV-2 virus has become a worldwide pandemic, and the number of patients presenting with respiratory failure is rapidly increasing in Japan. An international meta-analysis has been conducted to identify genetic factors associated with the onset and severity of COVID-19, but these factors have yet to be fully clarified. Here, we carried out genomic analysis based on a genome-wide association study (GWAS) in Japanese COVID-19 patients to determine whether genetic factors reported to be associated with the onset or severity of COVID-19 in the international meta-GWAS are replicated in the Japanese population, and whether new genetic factors exist. Although no significant genome-wide association was detected in the Japanese GWAS, an integrated analysis with the international meta-GWAS identified for the first time the involvement of the IL17A/IL17F gene in the severity of COVID-19. Among nine genes reported in the international meta-GWAS as genes involved in the onset of COVID-19, the association of FOXP4-AS1, ABO, and IFNAR2 genes was replicated in the Japanese population. Moreover, combined analysis of ABO and FUT2 genotypes revealed that the presence of oral AB antigens was significantly associated with the onset of COVID-19. FOXP4-AS1 and IFNAR2 were also significantly associated in the integrated analysis of the Japanese GWAS and international meta-GWAS when compared with severe COVID-19 cases and the general population. This made it clear that these two genes were also involved in not only the onset but also the severity of COVID-19. In particular, FOXP4-AS1 was not found to be associated with the severity of COVID-19 in the international meta-GWAS, but an integrated analysis with the Japanese GWAS revealed an association with severity. Individuals with the SNP risk allele found between IL17A and IL17F had significantly lower mRNA expression levels of IL17F, suggesting that activation of the innate immune response by IL17F may play an important role in the severity of SARS-CoV-2 infection.


Subject(s)
ABO Blood-Group System/genetics , COVID-19/pathology , Interleukin-17/genetics , Saliva/metabolism , Adult , Aged , Aged, 80 and over , Alleles , COVID-19/genetics , Female , Genome-Wide Association Study , Humans , Japan , Male , Middle Aged , Polymorphism, Single Nucleotide , Risk Factors , SARS-CoV-2/isolation & purification , Severity of Illness Index , Young Adult
9.
Int J Mol Sci ; 23(7)2022 Mar 28.
Article in English | MEDLINE | ID: covidwho-1785733

ABSTRACT

Patients with head and neck cancer (HNC) and patients with primary Sjögren's syndrome (pSS) may exhibit similar symptoms of dry mouth and dry eyes, as a result of radiotherapy (RT) or a consequence of disease progression. To identify the proteins that may serve as promising disease biomarkers, we analysed saliva and tears from 29 radiated HNC patients and 21 healthy controls, and saliva from 14 pSS patients by mass spectrometry-based proteomics. The study revealed several upregulated, and in some instances overlapping, proteins in the two patient groups. Histone H1.4 and neutrophil collagenase were upregulated in whole saliva of both patient groups, while caspase-14, histone H4, and protein S100-A9 were upregulated in HNC saliva only. In HCN tear fluid, the most highly upregulated protein was mucin-like protein 1. These overexpressed proteins in saliva and tears play central roles in inflammation, host cell injury, activation of reactive oxygen species, and tissue repair. In conclusion, the similarities and differences in overexpressed proteins detected in saliva from HNC and pSS patients may contribute to the overall understanding of the different pathophysiological mechanisms inducing dry mouth. Thus, the recurring proteins identified could possibly serve as future promising biomarkers.


Subject(s)
Head and Neck Neoplasms , Sjogren's Syndrome , Xerostomia , Biomarkers/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Histones/metabolism , Humans , Neoplasm Recurrence, Local/metabolism , Proteomics , Saliva/metabolism , Sjogren's Syndrome/metabolism , Tears/metabolism , Xerostomia/metabolism
10.
J Immunol ; 208(6): 1500-1508, 2022 03 15.
Article in English | MEDLINE | ID: covidwho-1715878

ABSTRACT

Oral fluids offer a noninvasive sampling method for the detection of Abs. Quantification of IgA and IgG Abs in saliva allows studies of the mucosal and systemic immune response after natural infection or vaccination. We developed and validated an enzyme immunoassay (EIA) to detect and quantify salivary IgA and IgG Abs against the prefusion-stabilized form of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein expressed in suspension-adapted HEK-293 cells. Normalization against total Ab isotype was performed to account for specimen differences, such as collection time and sample volume. Saliva samples collected from 187 SARS-CoV-2 confirmed cases enrolled in 2 cohorts and 373 prepandemic saliva samples were tested. The sensitivity of both EIAs was high (IgA, 95.5%; IgG, 89.7%) without compromising specificity (IgA, 99%; IgG, 97%). No cross-reactivity with endemic coronaviruses was observed. The limit of detection for SARS-CoV-2 salivary IgA and IgG assays were 1.98 ng/ml and 0.30 ng/ml, respectively. Salivary IgA and IgG Abs were detected earlier in patients with mild COVID-19 symptoms than in severe cases. However, severe cases showed higher salivary Ab titers than those with a mild infection. Salivary IgA titers quickly decreased after 6 wk in mild cases but remained detectable until at least week 10 in severe cases. Salivary IgG titers remained high for all patients, regardless of disease severity. In conclusion, EIAs for both IgA and IgG had high specificity and sensitivity for the confirmation of current or recent SARS-CoV-2 infections and evaluation of the IgA and IgG immune response.


Subject(s)
Antibodies, Viral/metabolism , COVID-19/diagnosis , Enzyme-Linked Immunosorbent Assay/methods , Immunoglobulin A/metabolism , Immunoglobulin G/metabolism , SARS-CoV-2/physiology , Saliva/metabolism , Adolescent , Adult , Aged , Asymptomatic Diseases , Child , Child, Preschool , Disease Progression , Enzyme-Linked Immunosorbent Assay/standards , Female , Humans , Infant , Male , Mass Screening , Middle Aged , Pandemics , Reference Standards , Sensitivity and Specificity , Severity of Illness Index , Young Adult
11.
PLoS One ; 17(2): e0263419, 2022.
Article in English | MEDLINE | ID: covidwho-1674014

ABSTRACT

Mucosal immunity plays a crucial role in controlling upper respiratory infections, including influenza. We established a quantitative ELISA to measure the amount of influenza virus-specific salivery IgA (sIgA) and salivary IgG (sIgG) antibodies using a standard antibody broadly reactive to the influenza A virus. We then analyzed saliva and serum samples from seven individuals infected with the A(H1N1)pdm09 influenza virus during the 2019-2020 flu seasons. We detected an early (6-10 days post-infection) increase of sIgA in five of the seven samples and a later (3-5 weeks) increase of sIgG in six of the seven saliva samples. Although the conventional parenteral influenza vaccine did not induce IgA production in saliva, vaccinated individuals with a history of influenza infection had higher basal levels of sIgA than those without a history. Interestingly, we observed sIgA and sIgG in an asymptomatic individual who had close contact with two influenza cases. Both early mucosal sIgA secretion and late systemically induced sIgG in the mucosal surface may protect against virus infection. Despite the small sample size, our results indicate that the saliva test system can be useful for analyzing upper mucosal immunity in influenza.


Subject(s)
Immunity, Mucosal/physiology , Influenza, Human/immunology , Saliva/immunology , Adult , Aged , Antibodies, Viral/analysis , Antibodies, Viral/metabolism , Antibody Formation , Cohort Studies , Female , History, 21st Century , Humans , Immunoglobulin A/analysis , Immunoglobulin A/metabolism , Immunoglobulin A, Secretory/analysis , Immunoglobulin A, Secretory/metabolism , Immunoglobulin G/analysis , Immunoglobulin G/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/therapeutic use , Influenza, Human/diagnosis , Influenza, Human/prevention & control , Japan , Longitudinal Studies , Male , Predictive Value of Tests , Prognosis , Saliva/chemistry , Saliva/metabolism , Young Adult
12.
Adv Mater ; 34(8): e2107892, 2022 Feb.
Article in English | MEDLINE | ID: covidwho-1634021

ABSTRACT

Surface chemistry critically affects the diagnostic performance of biosensors. An ideal sensor surface should be resistant to nonspecific protein adsorption, yet be conducive to analytical responses. Here a new polymeric material, zwitterionic polypyrrole (ZiPPy), is reported to produce optimal surface condition for biosensing electrodes. ZiPPy combines two unique advantages: the zwitterionic function that efficiently hydrates electrode surface, hindering nonspecific binding of hydrophobic proteins; and the pyrrole backbone, which enables rapid (<7 min), controlled deposition of ZiPPy through electropolymerization. ZiPPy-coated electrodes show lower electrochemical impedance and less nonspecific protein adsorption (low fouling), outperforming bare and polypyrrole-coated electrodes. Moreover, affinity ligands for target biomarkers can be immobilized together with ZiPPy in a single-step electropolymerization. ZiPPy-coated electrodes are developed with specificity for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The prepared sensor detects SARS-CoV-2 antibodies in human saliva down to 50 ng mL-1 , without the need for sample purification or secondary labeling.


Subject(s)
Antibodies, Viral/analysis , Biosensing Techniques/methods , COVID-19/diagnosis , Polymers/chemistry , Pyrroles/chemistry , Biosensing Techniques/instrumentation , COVID-19/virology , Electrochemical Techniques , Electrodes , Electroplating , Gold/chemistry , Humans , Limit of Detection , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , Saliva/metabolism , Surface Properties
13.
J Immunol ; 208(4): 819-826, 2022 02 15.
Article in English | MEDLINE | ID: covidwho-1638521

ABSTRACT

We used a noninvasive electrochemical quantitative assay for IgG Abs to SARS-CoV-2 S1 Ag in saliva to investigate the kinetics of Ab response in a community-based population that had received either the Pfizer or Moderna mRNA-based vaccine. Samples were received from a total of 97 individuals, including a subset of 42 individuals who collected samples twice weekly for 3 mo or longer. In all, >840 samples were collected and analyzed. In all individuals, salivary SARS-CoV-2 S1 IgG Ab levels rose sharply in the 2-wk period after their second vaccination, with peak Ab levels seen at 10-20 d after vaccination. We observed that 20%, 10%, and 2.4% of individuals providing serial samples had a 90%, 95%, and 99% drop, respectively, from peak levels during the duration of monitoring, and in two patients, Abs fell to prevaccination levels (5%). The use of noninvasive quantitative salivary Ab measurement can allow widespread, cost-effective monitoring of vaccine response.


Subject(s)
2019-nCoV Vaccine mRNA-1273/immunology , Antibodies, Viral/metabolism , BNT162 Vaccine/immunology , COVID-19/immunology , Immunoglobulin G/metabolism , SARS-CoV-2/physiology , Saliva/metabolism , Adult , Age Factors , Community-Based Participatory Research , Female , Humans , Male , Vaccination
14.
Sci Rep ; 11(1): 22493, 2021 11 18.
Article in English | MEDLINE | ID: covidwho-1526101

ABSTRACT

The COVID-19 pandemic caused by SARS-CoV-2 has infected millions worldwide, therefore there is an urgent need to increase our diagnostic capacity to identify infected cases. Although RT-qPCR remains the gold standard for SARS-CoV-2 detection, this method requires specialised equipment in a diagnostic laboratory and has a long turn-around time to process the samples. To address this, several groups have recently reported the development of loop-mediated isothermal amplification (LAMP) as a simple, low cost and rapid method for SARS-CoV-2 detection. Herein we present a comparative analysis of three LAMP-based assays that target different regions of the SARS-CoV-2: ORF1ab RdRP, ORF1ab nsp3 and Gene N. We perform a detailed assessment of their sensitivity, kinetics and false positive rates for SARS-CoV-2 diagnostics in LAMP or RT-LAMP reactions, using colorimetric or fluorescent detection. Our results independently validate that all three assays can detect SARS-CoV-2 in 30 min, with robust accuracy at detecting as little as 1000 RNA copies and the results can be visualised simply by color changes. Incorporation of RT-LAMP with fluorescent detection further increases the detection sensitivity to as little as 100 RNA copies. We also note the shortcomings of some LAMP-based assays, including variable results with shorter reaction time or lower load of SARS-CoV-2, and false positive results in some experimental conditions and clinical saliva samples. Overall for RT-LAMP detection, the ORF1ab RdRP and ORF1ab nsp3 assays have faster kinetics for detection but varying degrees of false positives detection, whereas the Gene N assay exhibits no false positives in 30 min reaction time, which highlights the importance of optimal primer design to minimise false-positives in RT-LAMP. This study provides validation of the performance of LAMP-based assays as a rapid, highly sensitive detection method for SARS-CoV-2, which have important implications in development of point-of-care diagnostics for SARS-CoV-2.


Subject(s)
COVID-19 Nucleic Acid Testing , COVID-19 , Molecular Diagnostic Techniques , Nucleic Acid Amplification Techniques , SARS-CoV-2/genetics , Saliva/metabolism , Adult , COVID-19/diagnosis , COVID-19/genetics , COVID-19/metabolism , Female , Humans , Male , Saliva/virology
15.
Mol Ther ; 30(5): 1966-1978, 2022 05 04.
Article in English | MEDLINE | ID: covidwho-1510414

ABSTRACT

To advance a novel concept of debulking virus in the oral cavity, the primary site of viral replication, virus-trapping proteins CTB-ACE2 were expressed in chloroplasts and clinical-grade plant material was developed to meet FDA requirements. Chewing gum (2 g) containing plant cells expressed CTB-ACE2 up to 17.2 mg ACE2/g dry weight (11.7% leaf protein), have physical characteristics and taste/flavor like conventional gums, and no protein was lost during gum compression. CTB-ACE2 gum efficiently (>95%) inhibited entry of lentivirus spike or VSV-spike pseudovirus into Vero/CHO cells when quantified by luciferase or red fluorescence. Incubation of CTB-ACE2 microparticles reduced SARS-CoV-2 virus count in COVID-19 swab/saliva samples by >95% when evaluated by microbubbles (femtomolar concentration) or qPCR, demonstrating both virus trapping and blocking of cellular entry. COVID-19 saliva samples showed low or undetectable ACE2 activity when compared with healthy individuals (2,582 versus 50,126 ΔRFU; 27 versus 225 enzyme units), confirming greater susceptibility of infected patients for viral entry. CTB-ACE2 activity was completely inhibited by pre-incubation with SARS-CoV-2 receptor-binding domain, offering an explanation for reduced saliva ACE2 activity among COVID-19 patients. Chewing gum with virus-trapping proteins offers a general affordable strategy to protect patients from most oral virus re-infections through debulking or minimizing transmission to others.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Angiotensin-Converting Enzyme 2/genetics , Animals , Chewing Gum , Cricetinae , Cricetulus , Cytoreduction Surgical Procedures , Humans , Protein Binding , SARS-CoV-2 , Saliva/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Virus Internalization
16.
Sci Rep ; 11(1): 21126, 2021 10 26.
Article in English | MEDLINE | ID: covidwho-1493210

ABSTRACT

Rapid identification of SARS-CoV-2-infected individuals is a cornerstone for the control of virus spread. The sensitivity of SARS-CoV-2 RNA detection by RT-PCR is similar in saliva and nasopharyngeal swabs. Rapid molecular point-of-care tests in saliva could facilitate, broaden and speed up the diagnosis. We conducted a prospective study in two community COVID-19 screening centers to evaluate the performances of a CE-marked RT-LAMP assay (EasyCoV) designed for the detection of SARS-CoV2 RNA from fresh saliva samples, compared to nasopharyngeal RT-PCR, to saliva RT-PCR and to nasopharyngeal antigen testing. Overall, 117 of the 1718 participants (7%) tested positive with nasopharyngeal RT-PCR. Compared to nasopharyngeal RT-PCR, the sensitivity and specificity of the RT-LAMP assay in saliva were 34% and 97%, respectively. The Ct values of nasopharyngeal RT-PCR were significantly lower in the 40 true positive subjects with saliva RT-LAMP (Ct 25.9) than in the 48 false negative subjects with saliva RT-LAMP (Ct 28.4) (p = 0.028). Considering six alternate criteria for reference tests, including saliva RT-PCR and nasopharyngeal antigen, the sensitivity of saliva RT-LAMP ranged between 27 and 44%. The detection of SARS-CoV-2 in crude saliva samples with an RT-LAMP assay had a lower sensitivity than nasopharyngeal RT-PCR, saliva RT-PCR and nasopharyngeal antigen testing.Registration number: NCT04578509.


Subject(s)
Ambulatory Care/methods , COVID-19 Nucleic Acid Testing/methods , COVID-19/diagnosis , COVID-19/metabolism , SARS-CoV-2 , Saliva/metabolism , Adult , Diagnostic Tests, Routine , False Negative Reactions , False Positive Reactions , Female , Humans , Male , Middle Aged , Molecular Diagnostic Techniques , Molecular Medicine , Nasopharynx/virology , Nucleic Acid Amplification Techniques , Point-of-Care Systems , Point-of-Care Testing , Prospective Studies , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Sensitivity and Specificity
17.
Emerg Microbes Infect ; 10(1): 1191-1199, 2021 Dec.
Article in English | MEDLINE | ID: covidwho-1246663

ABSTRACT

The ongoing COVID-19 pandemic has led to more than 159 million confirmed cases with over 3.3 million deaths worldwide, but it remains mystery why most infected individuals (∼98%) were asymptomatic or only experienced mild illness. The same mystery applies to the deadly 1918 H1N1 influenza pandemic, which has puzzled the field for a century. Here we discuss dual potential properties of the 1918 H1N1 pandemic viruses that led to the high fatality rate in the small portion of severe cases, while about 98% infected persons in the United States were self-limited with mild symptoms, or even asymptomatic. These variations now have been postulated to be impacted by polymorphisms of the sialic acid receptors in the general population. Since coronaviruses (CoVs) also recognize sialic acid receptors and cause severe acute respiratory syndrome epidemics and pandemics, similar principles of influenza virus evolution and pandemicity may also apply to CoVs. A potential common principle of pathogen/host co-evolution of influenza and CoVs under selection of host sialic acids in parallel with different epidemic and pandemic influenza and coronaviruses is discussed.


Subject(s)
COVID-19/pathology , Influenza, Human/pathology , Receptors, Cell Surface/genetics , Receptors, Virus/genetics , Sialic Acids/metabolism , Asymptomatic Diseases , Biological Evolution , COVID-19/mortality , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza, Human/mortality , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism , SARS-CoV-2/genetics , Saliva/metabolism , Saliva/virology
18.
Sci Rep ; 11(1): 710, 2021 01 12.
Article in English | MEDLINE | ID: covidwho-1242036

ABSTRACT

Saliva omics has immense potential for non-invasive diagnostics, including monitoring very young or elderly populations, or individuals in remote locations. In this study, multiple saliva omics from an individual were monitored over three periods (100 timepoints) involving: (1) hourly sampling over 24 h without intervention, (2) hourly sampling over 24 h including immune system activation using the standard 23-valent pneumococcal polysaccharide vaccine, (3) daily sampling for 33 days profiling the post-vaccination response. At each timepoint total saliva transcriptome and proteome, and small RNA from salivary extracellular vesicles were profiled, including mRNA, miRNA, piRNA and bacterial RNA. The two 24-h periods were used in a paired analysis to remove daily variation and reveal vaccination responses. Over 18,000 omics longitudinal series had statistically significant temporal trends compared to a healthy baseline. Various immune response and regulation pathways were activated following vaccination, including interferon and cytokine signaling, and MHC antigen presentation. Immune response timeframes were concordant with innate and adaptive immunity development, and coincided with vaccination and reported fever. Overall, mRNA results appeared more specific and sensitive (timewise) to vaccination compared to other omics. The results suggest saliva omics can be consistently assessed for non-invasive personalized monitoring and immune response diagnostics.


Subject(s)
Pneumococcal Infections/immunology , Pneumococcal Vaccines/administration & dosage , Proteome/drug effects , Saliva/metabolism , Sinusitis/immunology , Streptococcus pneumoniae/immunology , Transcriptome/drug effects , Adult , Humans , Immunity , Longitudinal Studies , Male , Pneumococcal Infections/drug therapy , Pneumococcal Infections/microbiology , Saliva/drug effects , Sinusitis/drug therapy , Sinusitis/microbiology , Time Factors , Vaccination
19.
Eur Rev Med Pharmacol Sci ; 25(8): 3154-3161, 2021 04.
Article in English | MEDLINE | ID: covidwho-1227268

ABSTRACT

OBJECTIVE: While both first-line antioxidant enzymes and oxidation products have been considered as markers of periodontal disease, their assessment in the diagnosis of periodontal disease is more complicated. Some, such as superoxide dismutase (SOD, glutathione peroxidase (GPx) and reduced glutathione (GSH), have indicated significant differences between patients with chronic and aggressive periodontitis. PATIENTS AND METHODS: Participants (101) were divided into a control group of healthy individuals and, following diagnosis, patients with gingivitis, chronic periodontitis, and aggressive periodontitis. Compounds reflecting tissue destruction, inflammatory processes or antioxidant responses, such as sirtuins (SIRT-1, SIRT-2), metalloproteinases (MMP), SOD, GPx, GSH, and glutathione reductase (GR) were measured in saliva. RESULTS: SIRT-2 levels were significantly increased in all patients. In patients with gingivitis, MMP (p<0.05) and GPx (p<0.01) were significantly increased. In patients with chronic and aggressive periodontitis, SOD activities were increased (p<0.001) while GPx and GR were decreased (p<0.001). Relative activities of MMP were higher in patients with aggressive periodontitis. CONCLUSIONS: Measurements of SIRT-2 and SOD clearly showed increased levels of oxidative stress in cases of periodontitis with a subsequent inhibition of other antioxidant enzymes. Levels of GSH suggest reversibility of the conditions with appropriate intervention. With the assessment of the trends of these selected antioxidant markers, it is possible to determine the prognosis of the disease.


Subject(s)
Periodontitis/metabolism , Saliva/metabolism , Sirtuin 2/metabolism , Superoxide Dismutase/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Humans , Periodontitis/diagnosis , Prognosis , Sirtuin 2/analysis , Superoxide Dismutase/analysis
20.
Encephale ; 46(3S): S35-S39, 2020 Jun.
Article in French | MEDLINE | ID: covidwho-1065051

ABSTRACT

OBJECTIVES: The ongoing COVID-19 pandemic comprises a total of more than 2,350,000 cases and 160,000 deaths. The interest in anti-coronavirus drug development has been limited so far and effective methods to prevent or treat coronavirus infections in humans are still lacking. Urgent action is needed to fight this fatal coronavirus infection by reducing the number of infected people along with the infection contagiousness and severity. Since the beginning of the COVID-19 outbreak several weeks ago, we observe in GHU PARIS Psychiatrie & Neurosciences (Sainte-Anne hospital, Paris, France) a lower prevalence of symptomatic and severe forms of COVID-19 infections in psychiatric patients (∼4%) compared to health care professionals (∼14%). Similar observations have been noted in other psychiatric units in France and abroad. Our hypothesis is that psychiatric patients could be protected from severe forms of COVID-19 by their psychotropic treatments. Chlorpromazine (CPZ) is a phenothiazine derivative widely used in clinical routine in the treatment of acute and chronic psychoses. This first antipsychotic medication has been discovered in 1952 by Jean Delay and Pierre Deniker at Sainte-Anne hospital. In addition, to its antipsychotic effects, several in vitro studies have also demonstrated a CPZ antiviral activity via the inhibition of clathrin-mediated endocytosis. Recently, independent studies revealed that CPZ is an anti-MERS-CoV and an anti-SARS-CoV-1 drug. In comparison to other antiviral drugs, the main advantages of CPZ lie in its biodistribution: (i) preclinical and clinical studies have reported a high CPZ concentration in the lungs (20-200 times higher than in plasma), which is critical because of the respiratory tropism of SARS-CoV-2; (ii) CPZ is highly concentrated in saliva (30-100 times higher than in plasma) and could therefore reduce the contagiousness of COVID-19; (iii) CPZ can cross the blood-brain barrier and could therefore prevent the neurological forms of COVID-19. METHODS: Our hypothesis is that CPZ could decrease the unfavorable evolution of COVID-19 infection in oxygen-requiring patients without the need for intensive care, but also reduce the contagiousness of SARS-CoV-2. At this end, we designed a pilot, phase III, multicenter, single blind, randomized controlled clinical trial. Efficacy of CPZ will be assessed according to clinical, biological and radiological criteria. The main objective is to demonstrate a shorter time to response (TTR) to treatment in the CPZ+standard-of-care (CPZ+SOC) group, compared to the SOC group. Response to treatment is defined by a reduction of at least one level of severity on the WHO-Ordinal Scale for Clinical Improvement (WHO-OSCI). The secondary objectives are to demonstrate in the CPZ+SOC group, compared to the SOC group: (A) superior clinical improvement; (B) a greater decrease in the biological markers of viral attack by SARS-CoV-2 (PCR, viral load); (C) a greater decrease in inflammatory markers (e.g. CRP and lymphopenia); (D) a greater decrease in parenchymal involvement (chest CT) on the seventh day post-randomization; (E) to define the optimal dosage of CPZ and its tolerance; (F) to evaluate the biological parameters of response to treatment, in particular the involvement of inflammatory cytokines. Patient recruitment along with the main and secondary objectives are in line with WHO 2020 COVID-19 guidelines. CONCLUSION: This repositioning of CPZ as an anti-SARS-CoV-2 drug offers an alternative and rapid strategy to alleviate the virus propagation and the infection severity and lethality. This CPZ repositioning strategy also avoids numerous developmental and experimental steps and can save precious time to rapidly establish an anti-COVID-19 therapy with well-known, limited and easy to manage side effects. Indeed, CPZ is an FDA-approved drug with an excellent tolerance profile, prescribed for around 70 years in psychiatry but also in clinical routine in nausea and vomiting of pregnancy, in advanced cancer and also to treat headaches in various neurological conditions. The broad spectrum of CPZ treatment - including antipsychotic, anxiolytic, antiemetic, antiviral, immunomodulatory effects along with inhibition of clathrin-mediated endocytosis and modulation of blood-brain barrier - is in line with the historical French commercial name for CPZ, i.e. LARGACTIL, chosen as a reference to its "LARGe ACTion" properties. The discovery of those CPZ properties, as for many other molecules in psychiatry, is both the result of serendipity and careful clinical observations. Using this approach, the field of mental illness could provide innovative therapeutic approaches to fight SARS-CoV-2.


Subject(s)
Antiviral Agents/therapeutic use , Betacoronavirus , Chlorpromazine/therapeutic use , Clinical Trials, Phase III as Topic/methods , Coronavirus Infections/drug therapy , Multicenter Studies as Topic/methods , Pandemics , Pneumonia, Viral/drug therapy , Randomized Controlled Trials as Topic/methods , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Biomarkers , Blood-Brain Barrier , COVID-19 , Chlorpromazine/pharmacokinetics , Chlorpromazine/pharmacology , Coronavirus Infections/epidemiology , Coronavirus Infections/immunology , Cytokines/blood , Dose-Response Relationship, Drug , Drug Repositioning , Endocytosis/drug effects , France/epidemiology , Humans , Lung/metabolism , Mental Disorders/drug therapy , Mental Disorders/epidemiology , Patient Selection , Pilot Projects , Pneumonia, Viral/epidemiology , Pneumonia, Viral/immunology , Research Design , SARS-CoV-2 , Saliva/metabolism , Severity of Illness Index , Single-Blind Method , Tissue Distribution , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL