Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
2.
Sci Rep ; 13(1): 7162, 2023 05 03.
Article in English | MEDLINE | ID: covidwho-2316264

ABSTRACT

Coronavirus disease (COVID-19) has generated interest in the assessment of systemic immune status, but existing knowledge about mucosal immunity is clearly insufficient to understand the full pathogenetic mechanisms of the disease. The aim of this study was to evaluate the long-term effects of novel coronavirus infection on mucosal immunity in the postinfection period among health care workers (HCWs). A total of 180 health care workers with and without a history of COVID-19 who ranged in age from 18 to 65 years were enrolled in this one-stage, cross-sectional study. The study subjects completed the 36-Item Short Form (36) Health Survey (SF-36) and the Fatigue Assessment Scale. Secretory immunoglobulin A (sIgA) and total immunoglobulin G (IgG) levels were quantified in saliva samples, induced sputum samples, and nasopharyngeal and oropharyngeal scrapings by an enzyme-linked immunosorbent assay. Specific anti-SARS-CoV-2 IgG antibodies were quantified in serum samples by chemiluminescence immunoassay. Analysis of the questionnaire data showed that all HCWs with a history of COVID-19 reported health problems that limited their daily activities and negative changes in their emotional health three months after the disease, regardless of its severity. The following shifts were detected in the adaptive arm of the immune response in different mucosal compartments. Among subjects who had severe or moderate-to-severe COVID-19, salivary sIgA levels were significantly higher than those in the control group (p < 0.05 and p < 0.005, respectively). Compared to the subjects in the control group, all subjects with prior COVID-19 had significantly higher levels of total IgG in induced sputum. In the group of patients who had had severe infection, total IgG in saliva was also higher (p < 0.05). A direct statistically significant correlation was also detected between the levels of total IgG in all studied samples and the levels of specific IgG antibodies against SARS-CoV-2 in the serum. A significant correlation was observed between total IgG levels and the parameters of physical and social activities, mental health, and fatigue levels. Our study demonstrated long-term changes in the humoral mucosal immune response, which were most pronounced in health care workers with a history of severe or moderate-to-severe COVID-19, and an association of these changes with certain clinical signs of post-COVID-19 syndrome.


Subject(s)
COVID-19 , Health Personnel , Immunity, Mucosal , Russia , COVID-19/immunology , COVID-19/pathology , COVID-19/physiopathology , Humans , Young Adult , Adult , Middle Aged , Immunoglobulin A/analysis , Respiratory System/immunology , Antibodies, Viral/analysis , Severity of Illness Index , Immunoglobulin G/analysis , SARS-CoV-2/physiology
3.
PeerJ ; 11: e15024, 2023.
Article in English | MEDLINE | ID: covidwho-2290901

ABSTRACT

Misdiagnosing suspected COVID-19 individuals could largely contribute to the viruses transmission, therefore, making an accurate diagnosis of infected subjects vital in minimizing and containing the disease. Although RT-PCR is the standard method in detecting COVID-19, it is associated with some limitations, including possible false negative results. Therefore, serological testing has been suggested as a complement assay to RT-PCR to support the diagnosis of acute infections. In this study, 15 out of 639 unvaccinated healthcare workers (HCWs) were tested negative for COVID-19 by RT-PCR and were found seropositive for SARS-CoV-2 nucleocapsid protein-specific IgM and IgG antibodies. These participants underwent additional confirmatory RT-PCR and SARS-CoV-2 spike-specific ELISA tests. Of the 15 individuals, nine participants were found negative by second RT-PCR but seropositive for anti-spike IgM and IgG antibodies and neutralizing antibodies confirming their acute infection. At the time of collection, these nine individuals were in close contact with COVID-19-confirmed patients, with 77.7% reporting COVID-19-related symptoms. These results indicate that including serological tests in the current testing profile can provide better outcomes and help contain the spread of the virus by increasing diagnostic accuracy to prevent future outbreaks rapidly.


Subject(s)
COVID-19 , Humans , COVID-19/diagnosis , SARS-CoV-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Immunoglobulin G/analysis , Immunoglobulin M/analysis , COVID-19 Testing
4.
Biosensors (Basel) ; 12(1)2021 Dec 23.
Article in English | MEDLINE | ID: covidwho-2250597

ABSTRACT

Sensitive serological assays are needed to provide valuable information about acute and past viral infections. For example, detection of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) IgG antibodies could serve as the basis for an "immunity passport" that would enable individuals to travel internationally. Here, utilizing a novel Magnetic Modulation Biosensing (MMB) system and the receptor-binding domain of the SARS-CoV-2 spike protein, we demonstrate a highly sensitive and specific anti-SARS-CoV-2 IgG serological assay. Using anti-SARS-CoV-2 IgG antibodies, RT-qPCR SARS-CoV-2-positive and healthy patients' samples, and vaccinees' samples, we compare the MMB-based SARS-CoV-2 IgG assay's analytical and clinical sensitivities to those of the enzyme-linked immunosorbent assay (ELISA). Compared with ELISA, the MMB-based assay has an ~6-fold lower limit of detection (129 ng/L vs. 817 ng/L), and it detects an increase in the IgG concentration much earlier after vaccination. Using 85 RT-qPCR SARS-CoV-2-positive samples and 79 -negative samples, the MMB-based assay demonstrated similar clinical specificity (98% vs. 99%) and sensitivity (93% vs. 92%) to the ELISA test, but with a much faster turnaround time (45 min vs. 245 min). The high analytical and clinical sensitivity, short turnaround time, and simplicity of the MMB-based assay makes it a preferred method for antibody detection.


Subject(s)
Antibodies, Viral/analysis , Biosensing Techniques , COVID-19 , Immunoglobulin G/analysis , Serologic Tests , COVID-19/diagnosis , COVID-19/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Magnetic Phenomena , SARS-CoV-2/immunology , Sensitivity and Specificity , Spike Glycoprotein, Coronavirus
5.
Environ Health Perspect ; 131(4): 47001, 2023 04.
Article in English | MEDLINE | ID: covidwho-2266850

ABSTRACT

BACKGROUND: Ambient air pollution has been associated with COVID-19 disease severity and antibody response induced by infection. OBJECTIVES: We examined the association between long-term exposure to air pollution and vaccine-induced antibody response. METHODS: This study was nested in an ongoing population-based cohort, COVICAT, the GCAT-Genomes for Life cohort, in Catalonia, Spain, with multiple follow-ups. We drew blood samples in 2021 from 1,090 participants of 2,404 who provided samples in 2020, and we included 927 participants in this analysis. We measured immunoglobulin M (IgM), IgG, and IgA antibodies against five viral-target antigens, including receptor-binding domain (RBD), spike-protein (S), and segment spike-protein (S2) triggered by vaccines available in Spain. We estimated prepandemic (2018-2019) exposure to fine particulate matter [PM ≤2.5µm in aerodynamic diameter (PM2.5)], nitrogen dioxide (NO2), black carbon (BC), and ozone (O3) using Effects of Low-Level Air Pollution: A Study in Europe (ELAPSE) models. We adjusted estimates for individual- and area-level covariates, time since vaccination, and vaccine doses and type and stratified by infection status. We used generalized additive models to explore the relationship between air pollution and antibodies according to days since vaccination. RESULTS: Among vaccinated persons not infected by SARS-CoV-2 (n=632), higher prepandemic air pollution levels were associated with a lower vaccine antibody response for IgM (1 month post vaccination) and IgG. Percentage change in geometric mean IgG levels per interquartile range of PM2.5 (1.7 µg/m3) were -8.1 (95% CI: -15.9, 0.4) for RBD, -9.9 (-16.2, -3.1) for S, and -8.4 (-13.5, -3.0) for S2. We observed a similar pattern for NO2 and BC and an inverse pattern for O3. Differences in IgG levels by air pollution levels persisted with time since vaccination. We did not observe an association of air pollution with vaccine antibody response among participants with prior infection (n=295). DISCUSSION: Exposure to air pollution was associated with lower COVID-19 vaccine antibody response. The implications of this association on the risk of breakthrough infections require further investigation. https://doi.org/10.1289/EHP11989.


Subject(s)
Air Pollutants , Air Pollution , COVID-19 , Humans , Air Pollutants/analysis , COVID-19 Vaccines , Spain , Antibody Formation , Environmental Exposure/analysis , SARS-CoV-2 , Air Pollution/analysis , Particulate Matter/analysis , Nitrogen Dioxide/analysis , Immunoglobulin G/analysis
6.
Orv Hetil ; 161(20): 807-812, 2020 05 01.
Article in Hungarian | MEDLINE | ID: covidwho-2224522

ABSTRACT

Introduction: At the end of March, 2020, rapid tests detecting the presence of antiviral IgM and IgG antibodies against SARS-CoV-2 virus were introduced in Hungary for the identification of SARS-CoV-2 infection (COVID-19 disease). Aim: We evaluated two rapid tests (Anhui and Clungene) in comparison with those of real-time PCR tests considered as the gold standard in the detection of infection. Method: Between 16, March and 14, April, 2020, we performed rapid IgM and IgG detecting tests without PCR; PCR without rapid tests; and PCR WITH rapid tests in 4140, 3210 and 1654 patients, respectively. (Out of these 1654 patients, Anhui and Clungene tests were used for testing in 625 and 1029 patients, respectively.) Patients were considered as positive in PCR and rapid tests when PCR positivity and IgM or IgG positivity occurred at any time, respectively. (Note: Clungene test is also marketed as 'Lungene'.) Results: The prevalence of PCR positivity in 4864 patients tested with PCR was 6.3%. The sensitivity and specificity of Anhui and Clungene tests were 33.3% and 72.85%, and 35.48% and 85.02%, respectively. At 6% PCR positivity, the positive and negative predictive values of Anhui and Clungene were 7.28%, 94.48%, 13.13%, and 95.38%, respectively. Conclusion: The low positive predictive values indicate that Anhui and Clungene rapid tests detecting the presence of anti-IgM and anti-IgG against SARS-CoV-2 virus infection are not suitable for screening SARS-CoV-2 vírus infection in the general population. These results strongly support that Anhui and Clungene rapid tests detecting IgM and IgG antibodies against SARS-CoV-2 virus should not be used in the differential diagnosis of infection. Orv Hetil. 2020; 161(20): 807-812.


Subject(s)
Antibodies, Viral , Coronavirus Infections/diagnosis , Immunoassay/methods , Pneumonia, Viral/diagnosis , Antibodies, Viral/analysis , Betacoronavirus , COVID-19 , COVID-19 Testing , Clinical Laboratory Techniques , Humans , Hungary , Immunoglobulin G/analysis , Immunoglobulin M/analysis , Pandemics , Predictive Value of Tests , Real-Time Polymerase Chain Reaction , SARS-CoV-2 , Sensitivity and Specificity
7.
BMC Infect Dis ; 23(1): 62, 2023 Feb 02.
Article in English | MEDLINE | ID: covidwho-2224135

ABSTRACT

BACKGROUND: Rapid IgM/IgG antibody tests were largely used in lieu of RT-PCR tests as part of COVID-19 public health response activities in Lima, Peru. To assess their utility, we explored the relationship between the time since onset of several COVID-19-related symptoms and the sensitivity of a rapid combined IgM/IgG antibody test. METHODS: We collected data from a community sample of individuals (n = 492) who received concurrent RT-PCR and rapid IgM/IgG antibody testing between May 2020 and March 2021. We estimated the sensitivity of the antibody test, against the RT-PCR test, by weeks since symptom onset via segmented regression analysis. RESULTS: The overall sensitivity of the rapid IgM/IgG antibody test was 46.7% (95% CI, 42.4-51.2%). Among 372 (75.6%) participants who reported COVID-19-related symptoms, sensitivity increased from 30.4% (95% CI, 24.7-36.6%) in week 1 after symptom onset to 83.3% (95% CI, 41.6-98.4%) in week 4. The test sensitivity increased by 31.9% (95% CI, 24.8-39.0%) per week until week 2 to 3, then decreased by - 6.0% (95% CI, - 25.7-13.7%) per week thereafter. CONCLUSION: Rapid antibody tests are a poor substitute for RT-PCR testing, regardless of presenting symptoms. This highlights the need for future pandemic planning to include timely and equitable access to gold-standard diagnostics, treatment, and vaccination.


Subject(s)
COVID-19 , Humans , COVID-19/diagnosis , SARS-CoV-2/genetics , Immunoglobulin G/analysis , Reverse Transcriptase Polymerase Chain Reaction , Peru/epidemiology , Sensitivity and Specificity , Immunoglobulin M/analysis , Antibodies, Viral/analysis , COVID-19 Testing
8.
Vaccine ; 40(48): 6963-6970, 2022 Nov 15.
Article in English | MEDLINE | ID: covidwho-2082524

ABSTRACT

BACKGROUND: The pandemic coronavirus disease 2019 (COVID-19) is a major global public health concern and several protective vaccines, or preventive/therapeutic approaches have been developed. Sinovac-CoronaVac, an inactivated whole virus vaccine, can protect against severe COVID-19 disease and hospitalization, but less is known whether it elicits long-term T cell responses and provides prolonged protection. METHODS: This is a longitudinal surveillance study of SARS-CoV-2 receptor binding domain (RBD)-specific IgG levels, neutralizing antibody levels (NAb), T cell subsets and activation, and memory B cells of 335 participants who received two doses of CoronaVac. SARS-CoV-2 RBD-specific IgG levels were measured by enzyme-linked immunosorbent assay (ELISA), while NAb were measured against two strains of SARS-CoV-2, the Wuhan and Delta variants. Activated T cells and subsets were identified by flow cytometry. Memory B and T cells were evaluated by enzyme-linked immune absorbent spot (ELISpot). FINDINGS: Two doses of CoronaVac elicited serum anti-RBD antibody response, elevated B cells with NAb capacity and CD4+ T cell-, but not CD8+ T cell-responses. Among the CD4+ T cells, CoronaVac activated mainly Th2 (CD4+ T) cells. Serum antibody levels significantly declined three months after the second dose. INTERPRETATION: CoronaVac mainly activated B cells but T cells, especially Th1 cells, were poorly activated. Activated T cells were mainly Th2 biased, demonstrating development of effector B cells but not long-lasting memory plasma cells. Taken together, these results suggest that protection with CoronaVac is short-lived and that a third booster dose of vaccine may improve protection.


Subject(s)
COVID-19 , Viral Vaccines , Humans , COVID-19/prevention & control , SARS-CoV-2 , COVID-19 Vaccines , Antibodies, Viral , Vaccination , Antibodies, Neutralizing , Immunoglobulin G/analysis , Th1 Cells , Vaccines, Inactivated
9.
Allergy Asthma Proc ; 43(5): 419-430, 2022 Sep 01.
Article in English | MEDLINE | ID: covidwho-2022489

ABSTRACT

Background: Secretory immunoglobulin A (sIgA) plays an important role in antiviral protective immunity. Although salivary testing has been used for many viral infections, including severe acute respiratory syndrome (SARS) and Middle East Respiratory Syndrome (MERS), its use has not yet been well established with the SARS coronavirus 2 (SARS-CoV-2). Quantification of salivary IgA and IgG antibodies can elucidate mucosal and systemic immune responses after natural infection or vaccination. Here, we report the development and validation of a rapid enzyme-linked immunosorbent assay (ELISA) for anti-SARS-CoV-2 salivary IgA and serum IgG antibodies, and present quantitative results for immunized subjects both prior to or following COVID-19 infections. Objective: Total and serum SARS-CoV-2 spike-specific IgG responses were compared with salivary spike-specific IgA and IgG responses in samples obtained from patients recently infected with SARS-CoV-2 and from subjects recently immunized with COVID-19 vaccines. Methods: A total of 52 paired saliva and serum samples were collected from 26 study participants: 7 subjects after COVID-19 infection and 19 subjects who were uninfected. The ELISA results from these samples were compared with five prepandemic control serum samples. Total IgG and SARS-CoV-2 spike-specific IgG in the serum samples from the subjects who were infected and vaccinated were also measured in a commercial laboratory with an enzyme immunoassay. Results: A wide variation in antibody responses was seen in salivary and serum samples measured by both methods. Three groups of serum total and IgG spike-specific SARS-CoV-2 antibody responses were observed: (1) low, (2) intermediate, and (3) high antibody responders. A correlational analysis of salivary IgA (sIgA) responses with serum IgG concentrations showed a statistical correlation in the low and intermediate antibody responder groups but not in the high group (which we believe was a result of saturation). Conclusion: These preliminary findings suggest measuring salivary and serum IgG and IgA merit further investigation as markers of current or recent SARS-CoV-2 infections.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunoglobulin A , Immunoglobulin G , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Antibodies, Viral , COVID-19/blood , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/immunology , Humans , Immunization , Immunoglobulin A/analysis , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin A, Secretory , Immunoglobulin G/analysis , Immunoglobulin G/blood , Immunoglobulin G/immunology , Saliva/chemistry , Saliva/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccination
10.
J Anim Sci ; 100(8)2022 Aug 01.
Article in English | MEDLINE | ID: covidwho-1890963

ABSTRACT

Passive transfer of colostral immunoglobulins from the cow to the calf is essential for calf health. The objective of this study was to determine if prepartum administration of a vaccine stimulates increased concentrations of colostral immunoglobulins of dairy cows beyond what is explained by vaccine-specific immunoglobulins. A prospective cohort study was conducted on a spring-calving commercial dairy farm that had a policy of only vaccinating cows with even ear tag numbers with a calf diarrhea vaccine, whereas cows with odd ear tag numbers were left unvaccinated. Cows in the vaccinated group (even ear tag numbers, n = 204) received a sensitizer and booster vaccination with a vaccine against bovine rotavirus (serotypes G6 and G10), bovine coronavirus, and E. coli having the K99 pili adherence factor. A sensitizer was given because the study vaccine was different from the vaccine previously used. Cows in the control group (odd ear tag numbers, n = 194) received a 2-mL subcutaneous sterile saline solution. Both groups received two treatments at a 3-wk interval, completing the treatments approximately 2 wk prior to the planned start of calving. During the calving period, technicians separated calves from cows immediately after parturition and prior to suckling, and cows were completely milked out within 6 h of parturition. Vaccine-specific, total, and nonvaccine-specific (total minus vaccine-specific) concentrations of immunoglobulin classes A, G1, G2a, and M (IgA, IgG1, IgG2a, and IgM, respectively) were quantified by mass spectrometry for 20 colostrum samples from each treatment group. Predicted mean non-vaccine-specific colostral IgM concentrations were 8.76 (95% CI = 7.18-10.67) and 5.78 (95% CI = 4.74-7.05) mg/mL for vaccinated and control cows, respectively (P = 0.005). Predicted mean non-vaccine-specific colostral IgG1 concentrations were 106.08 (95% CI = 92.07-120.08) and 95.30 (95% CI = 81.30-109.31) mg/mL among vaccinated and control cows, respectively; however, these means were not significantly different (P = 0.278). It is thus possible that the vaccine, in addition to specifically managing infectious calf diarrhea, may also have non-specific benefits by improving colostrum quality through increased non-vaccine-specific colostrum IgM concentrations. Further research is necessary to determine the mechanism for these preliminary findings, whether the effect may occur in other immunoglobulin classes, and what impacts it may have on calf health outcomes.


Unlike human babies, calves do not receive protective immune proteins (immunoglobulins) from the mother before birth, so a sufficient volume of immunoglobulin-rich colostrum of adequate quality must be consumed within hours of birth. It can be a challenge to meet this requirement for all dairy calves. Prior to calving, cows can be vaccinated with a vaccine against specific infectious causes of calf diarrhea to stimulate elevated concentrations of specific immunoglobulins in their colostrum, which is consumed by their calves to protect them until their own immune systems develop. We enrolled cows that were either vaccinated or not with a calf diarrhea vaccine and, using novel laboratory techniques, measured concentrations of immunoglobulin classes A, G, and M in their colostrum. As expected, vaccinated cows had elevated concentrations of vaccine-specific immunoglobulins in their colostrum. However, they also had elevated non-vaccine-specific concentrations of immunoglobulin M. The vaccine may therefore have stimulated a nonspecific increase in colostral immunoglobulin M concentrations. Further research is necessary to confirm the preliminary findings of the present study and determine the mechanism for this apparent nonspecific increase in colostral immunoglobulin M concentrations, whether it may occur in other immunoglobulin classes, and whether it may benefit calf health and growth.


Subject(s)
Colostrum , Vaccines , Animals , Animals, Newborn , Cattle , Colostrum/chemistry , Diarrhea/prevention & control , Diarrhea/veterinary , Escherichia coli , Female , Humans , Immunoglobulin G/analysis , Immunoglobulin M , Pregnancy , Prospective Studies
11.
Sci Rep ; 11(1): 24448, 2021 12 27.
Article in English | MEDLINE | ID: covidwho-1852474

ABSTRACT

COVID-19 mRNA vaccines are highly effective at preventing COVID-19. Prior studies have found detectable SARS-CoV-2 IgG antibodies in oral mucosal specimens of participants with history of COVID-19. To assess the development of oral SARS-CoV-2 IgG antibodies among people who received either the Moderna or Pfizer/BioNTech COVID-19 vaccination series, we developed a novel SARS-CoV-2 IgG enzyme-linked immunosorbent assay (ELISA) to quantify the concentrations of oral and nasal mucosal SARS-CoV-2 IgG levels. We enrolled 52 participants who received the Moderna vaccine and 80 participants who received the Pfizer/BioNTech vaccine. Oral mucosal specimens were self-collected by participants prior to or on the day of vaccination, and on days 5, 10, 15, and 20 following each vaccination dose and 30, 60, and 90 days following the second vaccination dose. A subset of the cohort provided additional nasal mucosal specimens at every time point. All participants developed detectable oral mucosal SARS-CoV-2 IgG antibodies by 15 days after the first vaccination dose. There were no significant differences in oral mucosal antibody concentrations once participants were fully vaccinated in the Moderna and Pfizer/BioNTech vaccines. Oral or nasal mucosal antibody testing could be an inexpensive and less invasive alternative to serum antibody testing. Further research is needed to understand the duration of detectable oral or nasal mucosal antibodies and how antibody concentrations change with time.


Subject(s)
Antibodies, Viral/analysis , Immunoglobulin G/analysis , Mouth Mucosa/metabolism , Respiratory System/metabolism , mRNA Vaccines/immunology , Adult , Aged , COVID-19/prevention & control , COVID-19/virology , Female , Health Personnel , Humans , Male , Middle Aged , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , Time Factors , Vaccination , Young Adult , mRNA Vaccines/administration & dosage
12.
Klin Lab Diagn ; 65(11): 688-692, 2020 Dec 04.
Article in English | MEDLINE | ID: covidwho-1780383

ABSTRACT

The study presents the results of the creation and evaluation of the diagnostic characteristics of the rapid immunochromatographic test for the qualitative detection and differentiation of IgM/IgG antibodies to SARS-CoV-2 in human serum, plasma, and whole blood "ИХА-COVID-19-IgM / IgG". Have been tested some samples without antibodies to SARS-CoV-2 and a samples with two and one type of specific antibodies. The coincidence of the results of immunochromatographic analysis with the results of the immunochemiluminescent method was 87.2%. Test kit can be use as the rapid diagnostic test in the context of the COVID-19 pandemic and to assess the immune status of convalescents.


Subject(s)
Antibodies, Viral/analysis , COVID-19 Serological Testing , COVID-19/diagnosis , Immunoassay , Immunoglobulin G/analysis , Immunoglobulin M/analysis , Humans
13.
Klin Lab Diagn ; 65(11): 683-687, 2020 Dec 04.
Article in English | MEDLINE | ID: covidwho-1780382

ABSTRACT

A new original Russian test kit for the detection of IgG-antibodies to the causative agent of COVID-19 - coronavirus SARS-CoV-2 by the method of enzyme-linked immunosorbent assay (ELISA) on a solid-phase «ELISA-SARS-CoV-2-AT-G¼ has been developed. In comparative tests with similar test systems «Vitrotest® SARS-CoV-2 IgG¼ (Vitrotest, Ukraine) and «Anti-SARS-Cov-2 ELISA (IgG)¼ (EUROIMMUN AG, Germany) high diagnostic efficiency of the new test system was shown.


Subject(s)
Antibodies, Viral/analysis , COVID-19 Serological Testing , COVID-19/diagnosis , Enzyme-Linked Immunosorbent Assay , Immunoglobulin G/analysis , Clinical Laboratory Techniques , Humans , Plasma , Reagent Kits, Diagnostic
14.
Obstet Gynecol ; 139(2): 181-191, 2022 02 01.
Article in English | MEDLINE | ID: covidwho-1774425

ABSTRACT

OBJECTIVE: To evaluate immune responses to coronavirus disease 2019 (COVID-19) mRNA-based vaccines present in breast milk and transfer of the immune responses to breastfeeding infants. METHODS: We enrolled 30 lactating women who received mRNA-based COVID-19 vaccines from January through April 2021 in this cohort study. Women provided serial milk samples, including milk expressed before vaccination, across 2-3 weeks after the first dose, and across 3 weeks after the second dose. Women provided their blood, spotted on cards (dried blood spots), 19 days after the first dose and 21 days after the second dose. Stool samples from the breastfed infants were collected 21 days after mothers' second vaccination. Prepandemic samples of milk, dried blood spots, and infant stool were used as controls. Milk, dried blood spots, and infant stool were tested by enzyme-linked immunosorbent assay for receptor-binding domain (RBD)-specific immunoglobulin (Ig)A and IgG. Milk samples were tested for the presence of neutralizing antibodies against the spike and four variants of concern: D614G, Alpha (B.1.1.7), Beta (B.1.351), and Gamma (P.1). Levels of 10 cytokines were measured in milk samples. RESULTS: Milk from COVID-19-immunized women neutralized the spike and four variants of concern, primarily driven by anti-RBD IgG. The immune response in milk also included significant elevation of interferon-γ. The immune response to maternal vaccination was reflected in breastfed infants: anti-RBD IgG and anti-RBD IgA were detected in 33% and 30% of infant stool samples, respectively. Levels of anti-RBD antibodies in infant stool correlated with maternal vaccine side effects. Median antibody levels against RBD were below the positive cutoffs in prepandemic milk and infant stool samples. CONCLUSION: Humoral and cellular immune responses to mRNA-based COVID-19 vaccination are present in most women's breast milk. The milk anti-RBD antibodies can neutralize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike and variants of concern. Anti-RBD antibodies are transferred to breastfed infants, with the potential to confer passive immunity against SARS-CoV-2.


Subject(s)
Antibodies, Neutralizing/analysis , Breast Feeding , COVID-19 Vaccines/immunology , Cytokines/analysis , Milk, Human/chemistry , SARS-CoV-2/immunology , Adult , Antibodies, Viral/analysis , Cohort Studies , Female , Humans , Immunoglobulin A/analysis , Immunoglobulin G/analysis , Infant , Infant, Newborn , Middle Aged , Vaccination
15.
Virol J ; 19(1): 24, 2022 02 03.
Article in English | MEDLINE | ID: covidwho-1770554

ABSTRACT

INTRODUCTION: In this work, six SARS-CoV-2-specific antibody assays were evaluated, namely, two pan-immunoglobulin (pan-Ig) assays [Roche Elecsys Anti-SARS-CoV-2 (named "Elecsys" in this study) and the PerkinElmer SuperFlex™ Anti-SARS-CoV-2 Ab Assay (SuperFlex_Ab)], two IgM assays [SuperFlex™ Anti-SARS-CoV-2 IgM Assay (SuperFlex_IgM) and YHLO iFlash-SARS-CoV-2 IgM (iFlash_IgM)], and two IgG assays [SuperFlex™ Anti-SARS-CoV-2 IgG Assay (SuperFlex_IgG) and iFlash-SARS-CoV-2 IgG (iFlash_IgG)]. Combination assays of SuperFlex™ (SuperFlex_any) and iFlash (iFlash_any) were also evaluated. METHODS: A total of 438 residual serum samples from 54 COVID-19 patients in the COVID-19 group and 100 samples from individuals without evidence of SARS-CoV-2 infection in the negative control group were evaluated. RESULTS: In the early stage of COVID-19 infection, within 14 days of symptom onset, the seropositive rate was lower than that of the late stage 15 days after onset (65.4% vs 99.6%). In the total period, the pan-Ig and IgG assays had higher sensitivity (90.8-95.3%) than the IgM assays (36.5-40.7%). SuperFlex_Ab and SuperFlex_any had higher sensitivity than Elecsys and SuperFlex_IgG (p < 0.05). The specificity of all the assays was 100%, except for SuperFlex_IgM (99.0%). The concordance rate between each assay was higher (96.4-100%) in the late stage than in the early stage (77.4-98.1%). CONCLUSION: For the purpose of COVID-19 diagnosis, antibody testing should be performed 15 days after onset. For the purpose of epidemiological surveillance, highly sensitive assays should be used as much as possible, such as SuperFlex_Ab, iFlash_IgG and their combination. IgM assays were not suitable for these purposes.


Subject(s)
Antibodies, Viral/analysis , COVID-19 Serological Testing/methods , COVID-19 , COVID-19/diagnosis , Humans , Immunoglobulin G/analysis , Immunoglobulin M/analysis , SARS-CoV-2/immunology , Sensitivity and Specificity
16.
Neurol Neuroimmunol Neuroinflamm ; 9(2)2022 03.
Article in English | MEDLINE | ID: covidwho-1745397

ABSTRACT

BACKGROUND AND OBJECTIVES: Information about humoral and cellular responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and antibody persistence in convalescent (COVID-19) patients with multiple sclerosis (PwMS) is scarce. The objectives of this study were to investigate factors influencing humoral and cellular responses to SARS-CoV-2 and its persistence in convalescent COVID-19 PwMS. METHODS: This is a retrospective study of confirmed COVID-19 convalescent PwMS identified between February 2020 and May 2021 by SARS-CoV-2 antibody testing. We examined relationships between demographics, MS characteristics, disease-modifying therapy (DMT), and humoral (immunoglobulin G against spike and nucleocapsid proteins) and cellular (interferon-gamma [IFN-γ]) responses to SARS-CoV-2. RESULTS: A total of 121 (83.45%) of 145 PwMS were seropositive, and 25/42 (59.5%) presented a cellular response up to 13.1 months after COVID-19. Anti-CD20-treated patients had lower antibody titers than those under other DMTs (p < 0.001), but severe COVID-19 and a longer time from last infusion increased the likelihood of producing a humoral response. IFN-γ levels did not differ among DMT. Five of 7 (71.4%) anti--CD20-treated seronegative patients had a cellular response. The humoral response persisted for more than 6 months in 41/56(81.13%) PwMS. In multivariate analysis, seropositivity decreased due to anti-CD20 therapy (OR 0.08 [95% CI 0.01-0.55]) and increased in males (OR 3.59 [1.02-12.68]), whereas the cellular response decreased in those with progressive disease (OR 0.04 [0.001-0.88]). No factors were associated with antibody persistence. DISCUSSION: Humoral and cellular responses to SARS-CoV-2 are present in COVID-19 convalescent PwMS up to 13.10 months after COVID-19. The humoral response decreases under anti-CD20 treatment, although the cellular response can be detected in anti-CD20-treated patients, even in the absence of antibodies.


Subject(s)
COVID-19/immunology , Immunity, Cellular , Immunity, Humoral , Multiple Sclerosis/immunology , Adult , Aged , Antibodies, Viral/analysis , Antigens, CD20/immunology , COVID-19/complications , Female , Humans , Immunoglobulin G/analysis , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Male , Middle Aged , Multiple Sclerosis/complications , Nucleocapsid/chemistry , Nucleocapsid/immunology , Retrospective Studies
17.
PLoS One ; 17(2): e0263468, 2022.
Article in English | MEDLINE | ID: covidwho-1702396

ABSTRACT

BACKGROUND: Mass vaccination is the key element in controlling current COVID-19 pandemic. Studies comparing immunogenicity of different COVID-19 vaccines are largely lacking. We aimed at measuring anti-S antibody (Ab) levels in individuals fully vaccinated with BNT162b2, BBIBP-CorV and Gam-COVID-Vac, as well as in COVID-19 convalescents. METHODS: In this cross-sectional study, serum was collected from 400 age- and sex-matched participants, 100 fully vaccinated with BNT162b2, 100 with BBIBP-CorV and 100 with Gam-COVID-Vac on the 28th day after the second vaccine dose, and 100 recovered from COVID-19 at least 28 days after symptom(s) resolution. Sera were analyzed using the LIAISON SARS-CoV-2 S1/S2 IgG assay (DiaSorin, Saluggia, Italy). Wilcoxon rank-sum or Kruskal-Wallis tests was used for comparison of Ab levels. RESULTS: Highest mean value (210.11, SD = 100.42) was measured in the BNT162b2 group, followed by Gam-COVID-Vac (171.11, SD = 120.69) and BBIBP-CorV (68.50, SD = 72.78) AU/mL (p<0.001). Significant differences in antibody levels were found between BNT162b2 and BBIBP-CorV (p<0.001), BNT162b2 and Gam-COVID-Vac (p = 0.001), as well as BBIBP-CorV and Gam-COVID-Vac groups (p<0.001). Percentage of seropositive was 81% in the convalescent group, 83% in BBIBP-CorV vaccinated and 100% in BNT162b2 and Gam-COVID-Vac. When comparing measured antibody levels in vaccinated to those in COVID-19 recovered, significantly higher antibody levels were found for vaccinated with BNT162b2 (p<0.001), and with Gam-COVID-Vac (p<0.001), while for BBIBP-CorV there was no statistically significant difference (p = 0.641). CONCLUSIONS: All three investigated vaccines, BNT162b2, BBIBP-CorV and Gam-COVID-Vac, provide robust immune response 28 days after the second dose of vaccine, in the majority of participants. All individuals vaccinated with BNT162b2 and Gam-COVID-Vac seroconverted, while in vaccinated with BBIBP-CorV and COVID-19 recovered seroconversion rates were lower. Although less potent compared to other two vaccines, immune response after BBIBP-CorV was similar to response measured in convalescents. Challenge still remains to examine dynamics and durability of immunoprotection.


Subject(s)
COVID-19 Vaccines/therapeutic use , COVID-19/immunology , COVID-19/therapy , Treatment Outcome , Adult , Antibodies/analysis , Antibodies/blood , Antibodies, Viral/blood , BNT162 Vaccine/immunology , COVID-19/blood , COVID-19 Vaccines/immunology , Convalescence , Cross-Sectional Studies , Female , Humans , Immunity/immunology , Immunity, Innate/immunology , Immunogenicity, Vaccine/immunology , Immunoglobulin G/analysis , Immunoglobulin G/blood , Male , Middle Aged , SARS-CoV-2/immunology , Serbia , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Inactivated/immunology , Vaccines, Synthetic/immunology
18.
Front Immunol ; 13: 751705, 2022.
Article in English | MEDLINE | ID: covidwho-1686480

ABSTRACT

COVID-19 affects children to a lesser extent than adults but they can still get infected and transmit SARS-CoV-2 to their contacts. Field deployable non-invasive sensitive diagnostic techniques are needed to evaluate the infectivity dynamics of SARS-CoV-2 in pediatric populations and guide public health interventions, particularly if this population is not fully vaccinated. We evaluated the utility of high-throughput Luminex assays to quantify saliva IgM, IgA and IgG antibodies against five SARS-CoV-2 spike (S) and nucleocapsid (N) antigens in a contacts and infectivity longitudinal study in 122 individuals (52 children and 70 adults). We compared saliva versus serum/plasma samples in infected children and adults diagnosed by weekly RT-PCR over 35 days (n=62), and those who consistently tested negative over the same follow up period (n=60), in the Summer of 2020 in Barcelona, Spain. Saliva antibody levels in SARS-CoV-2 RT-PCR positive individuals were significantly higher than in negative individuals and correlated with those measured in sera/plasmas. Asymptomatic infected individuals had higher levels of anti-S IgG than symptomatic individuals, suggesting a protective anti-disease role for antibodies. Higher anti-S IgG and IgM levels in serum/plasma and saliva, respectively, in infected children compared to infected adults could also be related to stronger clinical immunity in them. Among infected children, males had higher levels of saliva IgG to N and RBD than females. Despite overall correlation, individual clustering analysis suggested that responses that may not be detected in blood could be patent in saliva, and vice versa. In conclusion, measurement of SARS-CoV-2-specific saliva antibodies should be considered as a complementary non-invasive assay to serum/plasma to determine COVID-19 prevalence and transmission in pediatric populations before and after vaccination campaigns.


Subject(s)
Antibodies, Viral/analysis , COVID-19 Serological Testing/methods , COVID-19/diagnosis , Immunoassay/methods , Saliva , Adult , Child , Female , Humans , Immunoglobulin A/analysis , Immunoglobulin G/analysis , Immunoglobulin M/analysis , Male , SARS-CoV-2 , Spain
19.
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
20.
PLoS One ; 17(2): e0263327, 2022.
Article in English | MEDLINE | ID: covidwho-1662444

ABSTRACT

Rapid screening and diagnosis of coronavirus disease 2019 in the emergency department is important for controlling infections. When polymerase chain reaction tests cannot be rapidly performed, rapid antigen testing is often used, albeit with insufficient sensitivity. Therefore, we evaluated the diagnostic accuracy of combining rapid antigen and antibody test results. This was a retrospective review of patients who visited our emergency department between February and May 2021 and underwent rapid antigen, immunoglobulin G antibody, and reverse transcription-polymerase chain reaction tests. The study included 1,070 patients, of whom 56 (5.2%) tested positive on reverse transcription-polymerase chain reaction. The sensitivity, specificity, and area under the curve of rapid antigen testing were 73.7%, 100.0%, and 0.87, respectively. The combined rapid antigen and antibody test result had improved diagnostic accuracy, with 91.2% sensitivity, 97.9% specificity, and an area under the curve of 0.95. The results of the rapid antigen and antibody tests could be combined as a reliable alternative to reverse transcription-polymerase chain reaction.


Subject(s)
COVID-19 Testing/methods , COVID-19/diagnosis , Point-of-Care Testing , Aged , Aged, 80 and over , COVID-19 Serological Testing/methods , Female , Humans , Immunoglobulin G/analysis , Male , Middle Aged , Retrospective Studies , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL