Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Atemwegs- und Lungenkrankheiten ; 49(4):129-133, 2023.
Article in German | EMBASE | ID: covidwho-20242600

ABSTRACT

The coronavirus SARS-CoV-2 was detected in isolates of pneumonia patients in January 2020. The virus cannot multiply extracellularly but requires access to the cells of a host organism. SARS-CoV-2 uses angiotensin-converting enzyme 2 (ACE2) as a receptor, to which it docks with its spikes. ACE2 belongs to the renin angiotensin system (RAS), whose inhibitors have been used for years against high blood pressure. Renin is an endopeptidase that is predominantly formed in the juxtaglomerular apparatus of the kidney and cleaves the decapeptide angiotensin I (Ang I) from angiotensinogen. Through the angiotensin-converting enzyme (ACE), another 2 C-terminal amino acids are removed from Ang I, so that finally the active octapeptide angiotensin II (Ang II) is formed. The biological effect of Ang II via the angiotensin II receptor subtype 1 (AT1-R) consists of vasoconstriction, fibrosis, proliferation, inflammation, and thrombosis formation. ACE2 is a peptidase that is a homolog of ACE. ACE2 is predominantly expressed by pulmonary alveolar epithelial cells in humans and has been detected in arterial and venous endothelial cells. In contrast to the dicarboxy-peptidase ACE, ACE2 is a monocarboxypeptidase that cleaves only one amino acid from the C-terminal end of the peptides. ACE2 can hydrolyze the nonapeptide Ang-(1-9) from the decapeptide Ang I and the heptapeptide Ang-(1-7) from the octapeptide Ang II. Ang-(1-7) acts predominantly antagonistically (vasodilatory, anti-fibrotic, anti-proliferative, anti-inflammatory, anti-thrombogenetically) via the G protein-coupled Mas receptor to the AT1-R-mediated effects of Ang II. In the pathogenesis of COVID-19 infection, it is therefore assumed that there is an imbalance due to overstimulation of the AT1 receptor in conjunction with a weakening of the biological effects of the Mas receptor.Copyright © 2022 Dustri-Verlag Dr. K. Feistle.

2.
American Journal of Reproductive Immunology ; 89(Supplement 1):54-55, 2023.
Article in English | EMBASE | ID: covidwho-20238235

ABSTRACT

Problem: Although it is rare for a SARS-CoV-2 infection to transmit vertically to the fetus during pregnancy, there is a significantly increased risk of adverse pregnancy outcomes due to maternalCOVID- 19. However, there is a poor understanding of such risks because mechanistic studies on how SARS-CoV-2 infection disrupts placental homeostasis are significantly lacking. The SARS-CoV-2 proteome includes multiple structural and non-structural proteins, including the non-structural accessory proteinORF3a. The roles of these proteins in mediating placental infection remain undefined. We and others have shown that autophagy activity in placental syncytium is essential for barrier function and integrity. Here, we have used clinical samples and cultured trophoblast cells to evaluate syncytial integrity of placenta exposed to SARS-CoV-2. The objective of our study was to investigate potential mechanisms through which SARS-CoV-2 impairs placental homeostasis and causes adverse pregnancy outcomes. We tested the central hypothesis that an essential SARS-CoV-2 non-structural and accessory protein, ORF3a, uniquely (amongst multiple viral proteins tested) with a novel three-dimensional structure andwith no homology to any other proteins is a key modulator of placental trophoblast cell dynamics via autophagy and intracellular trafficking of a tight junction protein (TJP), ZO-1. Method(s): We used clinical samples and cultured trophoblast cells to evaluate syncytial integrity of placentas exposed to SARS-CoV- 2. Autophagic flux was measured in placental villous biopsies from SARS-CoV-2-exposed and unexposed pregnant women by quantifying the expression of autophagy markers, LC3 and P62. Trophoblast cells (JEG-3, Forskolin-treated JEG-3, HTR8/SVneo, or primary human trophoblasts (PHTs)) were transfected with expression plasmids encoding SARS-CoV-2 proteins including ORF3a. Using western blotting, multi-label immunofluorescence, and confocal imaging, we analyzed the effect of ORF3a on the autophagy, differentiation, invasion, and intracellular trafficking of ZO-1 in trophoblasts. Using coimmunoprecipitation assays, we tested ORF3a interactions with host proteins. t-tests and one-way analyses of variance (ANOVAs) with post hoc tests were used to assess the data, with significance set at P < .05. Result(s): We discovered :1) increased activation of autophagy, but incomplete processing of autophagosome-lysosomal degradation;2) accumulation of protein aggregates in placentas exposed to SARS-CoV- 2. Mechanistically, we showed that the SARS-CoV-2 ORF3a protein, uniquely 3) blocks the autophagy-lysosomal degradation process;4) inhibits maturation of cytotrophoblasts into syncytiotrophoblasts (STBs);5) reduces production ofHCG-beta, a key pregnancy hormone that is also essential for STB maturation;and 6) inhibits trophoblast invasive capacity. Furthermore, ORF3a harbors an intrinsically disordered C-terminus withPDZ-bindingmotifs.We show for the first time that, 7) ORF3a binds to and co-localizes with the PDZ domain of ZO-1, a junctional protein that is essential for STB maturation and the integrity of the placental barrier. Conclusion(s): Our work outlines a new molecular and cellular mechanism involving the SARS-CoV-2 accessory protein ORF3a that may drive the virus's ability to infect the placenta and damage placental syncytial integrity. This implies that the mechanisms facilitating viral maturation, such as the interaction of ORF3a with host factors, can be investigated for additional functionality and even targeted for developing new intervention strategies for treatment or prevention of SARS-CoV-2 infection at the maternal-fetal interface.

3.
Topics in Antiviral Medicine ; 31(2):141, 2023.
Article in English | EMBASE | ID: covidwho-2319964

ABSTRACT

Background: More than 12 billion doses of COVID-19 vaccine administrations and over 630 million natural infections should have developed adequate levels of herd immunity over the last three years. However, there have been many new waves of coronavirus infections. The development of safe and effective vaccines to control breakthrough SARS-CoV-2 infections remain an urgent priority. We have developed a recombinant VSV vector-based vaccine to fulfill this worldwide need. Method(s): We have used a recombinant vesicular stomatitis virus (rVSV)-based prime-boost immunization strategy to develop an effective COVID-19 recall vaccine candidate. We have constructed an attenuated recombinant VSV genome carrying the full-length Spike protein gene of SARS-CoV-2. Adding the honeybee melittin signal peptide (msp) at the N-terminus enhanced the protein expression and adding the VSV G protein transmembrane domain and the cytoplasmic tail (Gtc) at the C-terminus of the Spike protein allowed efficient incorporation of the Spike protein into pseudotype VSV. Result(s): In immunized mice, rVSV with chimeric rVSV-msp-S-Gtc induced high levels of potent neutralizing antibodies (nAbs) and CD8+ T cell responses, while the full-length Spike with Gtc proved to be the superior immunogen. More importantly, rVSV-msp-S-Gtc-vaccinated animals were completely protected from subsequent SARS-CoV-2 challenges. Furthermore, rVSV-Wuhan and rVSV-Delta vaccines, and an rVSV-Trivalent (mixed rVSV-Wuhan, -Beta and -Delta) vaccine elicited potent nAbs against live SARS-CoV-2 Wuhan (USAWA1), Beta (B.1.351), Delta (B.1.617.2) and Omicron (B.1.1.529) viruses. Heterologous boosting of rVSV-Wuhan with rVSV-Delta induced strong nAb responses against Delta and Omicron viruses, with the rVSV-Trivalent vaccine consistently inducing effective nAbs against all the SARS-CoV-2 variants tested. All rVSV-msp-S-Gtc vaccines also elicited an immunodominant Spike-specific CD8+ T cell response. Conclusion(s): rVSV vaccines targeting SARS-CoV-2 variants of concern can be considered as an effective booster vaccine in the global fight against COVID-19.

4.
Acta Veterinaria et Zootechnica Sinica ; 53(7):2260-2267, 2022.
Article in Chinese | EMBASE | ID: covidwho-2245652

ABSTRACT

The C-terminal domain (CTD) of porcine deltacoronavirus S1 subunit is the main region which induces the neutralizing antibody. S1-CTD was expressed by HEK-293T eukaryotic expression system and purified, and porcine ileal epithelium cells membrane proteins were extracted to investigate porcine host proteins that interact with it. Thirty-two suspected interacting host proteins were obtained by co-inmunprecipitation (Co-IP) and mass spectrometry. Eukaryotic expression plasmid of KIF1 binding protein (KIFBP) was constructed, and the interaction between KIFBP and S1-CTD was identified by Co-IP and laser confocal microscopy. All results proved that KIFBP interacted with S1-CTD and co-located in cytoplasm. Further research indicated that overexpression of KIFBP could effectively reduce the viral mRNA level and the viral titer in which the mRNA level decreased by about 70%, and the viral titer decreased by 101.6TCID50. In conclusion, a host protein KIFBP interacting with PDCoV S1-CTD was screened and identified in this study which provides a theoretical basis for understanding the pathogenesis of PDCoV.

5.
Multiple Sclerosis Journal ; 28(3 Supplement):103-104, 2022.
Article in English | EMBASE | ID: covidwho-2138835

ABSTRACT

Introduction: The new SARS-CoV-2-mRNA-vaccines provide protection against severe COVID-19 infection. Disease modifying therapies (DMTs) for treatment of persons with multiple sclerosis (pwMS) differently impact humoral and cellular immunity and therefore can diminish vaccination outcomes. Thus, it is crucial to investigate the influence of different DMTs on the immune response after SARS-CoV-2 vaccination of pwMS. Objective(s): To investigate antibody and T-cell responses after SARS-CoV-2-vaccination in pwMS treated with different DMTs. Method(s): We studied antibody and T-cell responses in pwMS 4 and 12 weeks after the second dose of mRNA-vaccination against SARS-CoV-2. The results were compared to baseline samples taken before the first dose of SARS-CoV-2-vaccination. We screened and included 148 pwMS treatedwith natalizumab (n=23), dimethylfumarate (n=24), fingolimod (n=39), cladribine (n=31), alemtuzumab (n=17) and teriflunomide (n=14). Healthy controls (HC) (n=43) were used as a comparison. To evaluate humoral immune responses, IgG reactivity was measured towards three different SARS-CoV-2 antigens using a multiplex bead assay: full-length spike glycoprotein (spike S1S2 foldon), spike S1 domain and the nucleocapsid protein C-terminal domain (Nucleocapsid C). Furthermore, the antibody data allowed us to distinguish pwMS who had been vaccinated after a previous SARS-CoV-2-infection. Cellular immune responses were studied using a Fluorospot assay measuring IFNy and IL-13 T-cell responses to the spike S1 domain and Nucleocapsid C. Result(s): Humoral responses to vaccination were comparable between HC and pwMS treated with natalizumab, dimethylfumarate, cladribine, alemtuzumab and teriflunomide, but suppressed with fingolimod. In addition, T-cell responses were nearly absent in the fingolimod group and moderately reduced in the cladribine group. Conclusion(s): In this comprehensive study of both antibody and cellular responses to SARS-CoV-2-vaccination in pwMS on different DMTs, fingolimod was associated with abrogated responses in both aspects, while cladribine-treated individuals displayed reduced cellular response only. These findings are of relevance for risk mitigation strategies and vaccination recommendations for pwMS.

6.
Journal of the American Society of Nephrology ; 33:930, 2022.
Article in English | EMBASE | ID: covidwho-2126097

ABSTRACT

Introduction: Critically-ill patients with prolonged immobility are at risk of hypercalcemia. In ESRD patients with hypercalcemia therapeutic options are limited: Hydration may cause volume overload, loop diuretics are not effective and bisphosphonates may cause over suppression of bone turnover. This case series presents two patients with immobilization-induced hypercalcemia successfully treated with denosumab Case Description: Case 1: a 51 y/o male s/p bilateral lung transplant who developed chronic respiratory failure due to COVID-19. Case 2: a 54 y/o male initially admitted with COVID-19 requiring b/l lung transplant. Both patients developed AKI that progressed to ESRD and were bed bound. Evaluation for the hypercalcemia was compatible with immobilization-induced hypercalcemia: low iPTH, normal 1,25-VitD levels, negative multiple myeloma workup and mildly elevated PTH-rp (due to accumulation of the carboxy-terminal fragments). We eliminated calcium in the enteral feeds, used low calcium dialysis bath without success. Both patients received denosumab 60mg S/c once with improvement of the hypercalcemia. They developed asymptomatic hypocalcemia treated with resumption of calcium in the diet and using a 2.5Ca dialysis bath. Discussion(s): Denosumab is monoclonal antibody to the RANKL that inhibits osteoclast formation. It is currently used in osteoporosis therapy and cancer-induced hypercalcemia. In ESRD patients with immobilization-related hypercalcemia, denosumab offers the advantage of no renal dose adjustment necessary, rapid onset of action and longer therapeutic duration. Patient should be monitored for hypocalcemia, rebound hypercalcemia and risk of over suppression of bone remodeling. (Figure Presented).

7.
Annals of the Rheumatic Diseases ; 81:368-369, 2022.
Article in English | EMBASE | ID: covidwho-2008986

ABSTRACT

Background: In line with other reports, our group showed that patients treated with rituximab had signifcant impaired antibody response compared to patients treated with other biologic and targeted and synthetic disease modifying anti-rheumatic drugs (csDMARD). Objectives: To investigate predictors of response to COVID-19 vaccination (2 doses of mRNA vaccines, 2 doses of virus vector vaccines or combinations of these) in patients with infammatory rheumatic diseases (IRD) treated with ritux-imab and controls. Methods: Antibody levels to three antigens: Spike protein full length, Spike S1 and Nucleocapsid C-terminal fragment (to confrm previous COVID-19 infection) were measured in sera collected before vaccination and 2-12 weeks after the second vaccine using a multiplex bead-based serology assay. The antigen-specifc cut-off was defned as the median fuorescence intensity signal plus 6x standard deviations across 12 pre-pandemic controls. A good vaccine response was defned as having antibodies over the cut-off level for both spike antigens. Proportion (%) responders was compared between patients and controls (Chi2 test). Patients with IRD receiving last rituximab treatment within a mean (range) 193 (23-501) days before frst vaccination participated. Individuals without IRD served as a control group. Predictors of a good vaccine response were explored using multivariate logistic regression analysis adjusted for age, sex, disease duration, diagnosis (systemic vasculitis/RA/JIA/other), concomitant csDMARD, rituximab dose and prednisolone dose. Hazard ratio (chanse) of a good antibody response in relation to time between the last rituximab treatment and vaccination was studied by Kaplan-Meier survival analysis. Results: In total, 145 patients receiving rituximab and 61 controls were inclyded. Of these, 82 received rituximab as monotherapy (67% women;mean age 66 years, mean disease duration 13 years;33% had RA/JIA and 60% vasculi-tis) and 63 received rituximab+csDMARD (62% women;mean age 66 years;mean disease duration 17 years;76% had RA/JIA and 10 % vasculitis). Controls (n=61) were 74% women and mean age 49 years. Compared to controls, rituximab patients had lower antibody levels for both spike proteins (p<0.001). Proportion (%) responders among patients receiving rituximab as monotherapy (40.2%) and rituximab+DMARDs (25.4%) was signifcantly lower than in controls (98.4%) (p<0.001, Chi2). Higher age, concomitant csDMARD at vaccination and shorter time from last rituximab treatment predicted impaired antibody response (multivariate logistic regression model) (Table 1). Longer time between the last rituximab course and vaccination was associated with better antibody response (Figure 1). Conclusion: Patients with IRD getting vaccinated with two doses of COVID19 vaccine during the treatment with rituximab have the ability to develop antibody response although the response is impaired. For each month passed after the last rituximab course, the chance of good antibody response increases with 30%. Younger patients receiving rituximab as monotherapy and vaccinated preferably several months after the last rituximab treatment have the highest chance of achieving a good antibody response.

8.
Annals of the Rheumatic Diseases ; 81:113-114, 2022.
Article in English | EMBASE | ID: covidwho-2008896

ABSTRACT

Background: Initial studies on the immunogenicity of COVID-19 vaccines in patients with immune-mediated infammatory rheumatic diseases (IRD) reported diminished antibody response in general, and particularly when treated with rituximab or abatacept (1). Additional data are needed, especially for patients with IRD and immunomodulatory treatments. Objectives: To elucidate the antibody response after two doses of COVID-19 vaccine in patients with IRD treated with biologic or targeted synthetic disease modifying anti-rheumatic drugs (b/ts DMARDs) as monotherapy or combined with conventional synthetic DMARDS (csDMARDs). Methods: Antibodies against two antigens representing Spike full length protein and Spike S1 and a Nucleocapsid C-terminal fragment (used to confrm previously COVID-19 infection) were measured in serum obtained before and after the second vaccination using a multiplex bead-based serology assay (2). Patients with IRD receiving immunomodulating treatment, followed at a rheumatology department and healthy individuals (controls) were recruited from five Swedish regions. Antibody positivity was classifed as the signal passing an antigen specifc cutoff based on the mean intensity signal of 12 selected negative pre-pandemic controls plus 6SD for Spike/S1 and 12SD for Nucleocapsid-C. Good vaccine response was defned as having antibodies over cut-off level for both spike antigens. Percentage of responders in each treatment group was compared to controls (Chi2 test). Predictors of antibody response were determined using logistic regression analysis. Results: In total, 414 patients (320 RA/JIA/psoriatic arthritis/axial spondylar-thritis, 60 systemic vasculitis and 32 other IRD) and 61 controls participated. Patients receiving rituximab (n=145;65% female;mean age 65years), abatacept (n=21;77% female;mean age 66 years), IL6 inhibitors (n=77;74% female;mean age 64years), JAK-inhibitors (n=58;75% female, mean age 53years), TNF-inhib-itors (n=68;66% female;mean age 44years;), IL17 inhibitors (n=42;54% female;mean age 44years) and controls (n=61;74% female, mean age 49years) were studied. Patients receiving IL6 inhibitor (81.0%), abatacept (43.8%) or rituximab (33.8%) had a signifcantly lower antibody response rate compared to controls (98.4%), further pronounced if combined with csDMARD (p<0.001) (Figure 1). In the adjusted logistic regression analysis, higher age, rituximab, abatacept, concomitant csDMARD but not IL6 inhibitors, concomitant prednisolone, or a vascu-litis diagnosis, remained signifcant predictors of antibody response (Table 1). All vaccines were well tolerated. 14 (3.4%) patients reported an increased activity in their IRD following vaccination. Conclusion: In this nationwide study including IRD patients receiving b/ts DMARDs a decreased immunogenicity of COVID-19 vaccines was observed in patients receiving rituximab, abatacept and to some extent IL-6 inhibitors. Concomitant csDMARD gave further attenuation. Patients on rituximab and abata-cept should be prioritized for booster doses of COVID19 vaccine.

9.
FEBS Open Bio ; 12:287, 2022.
Article in English | EMBASE | ID: covidwho-1976659

ABSTRACT

The global pandemic prompted by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has already caused more than 6 million deaths worldwide, calling for urgent effective therapeutic measures. A deep understanding of the mechanisms involved in viral replication is required. Among the nonstructural proteins (nsps) encoded by SARS-CoV-2 genome, there is the nsp14 ribonuclease, the main object of study in this work. Ribonucleases are key factors in the control of all biological processes, ensuring maturation, degradation, and quality control of all types of RNAs. Nsp14 is a bifunctional protein, holding a 3'- 5' exoribonucleolytic activity (ExoN) in the N-terminal domain, stimulated through the interaction with nsp10, and a C-terminal N7-methyltransferase activity (MTase). Both are critical for the coronavirus life cycle. In this work, we provide a complete biochemical characterization of SARS-CoV-2 nsp14-nsp10, addressing several aspects of the complex for the first time. Moreover, using a homology model, we have identified residues involved in the nsp14-nsp10 interaction that were extensively studied. We have confirmed the SARS-CoV-2 nsp14 dual function and we have shown that both ExoN and MTase activities are functionally independent. We demonstrate that the nsp14 MTase activity is independent of nsp10, contrarily to nsp14 ExoN that is upregulated in the presence of the cofactor. Additionally, our results show that the ExoN motif I has a prominent role on the ribonucleolytic activity of SARS-CoV-2 nsp14, contrasting to what was previously observed in other coronaviruses, which can be related to the pathogenesis of SARS-CoV-2. The knowledge provided in this work can serve as a basis to design effective drugs that target the pinpointed residues in order to disturb the complex assembly and affect the viral replication, ultimately, treating COVID-19 and other CoV infections.

10.
Topics in Antiviral Medicine ; 30(1 SUPPL):93, 2022.
Article in English | EMBASE | ID: covidwho-1880277

ABSTRACT

Background: Following natural infection or vaccination, the generation of stem cell-like memory T (Tscm) cells is essential for long-term protective immunity to the virus. Tscm cells have the capacity for self-renewal and multipotency. In SARS-CoV2 infection, the emergence of CD8+ Tscm cells is correlated with the number of symptom-free days. The development of a COVID-19 vaccine able to generate CD8+ Tscm cells is of the utmost importance since the emergence of SARS-CoV2 variants of concerns requires maintaining strong and long-lasting immune responses, 2) as an efficient alternative in immunocompromised people who have difficulties raising humoral immune responses. Methods: We have developed a new Dendritic Cell-based vaccine composed of a humanized αCD40 monoclonal antibody fused to the RBD protein in its C-terminal Fc-domains and three T cell epitopes spanning sequences from S and N proteins in its light chains (αCD40-CoV2). Previous studies have shown that this platform elicited durable and robust T-and B-cell responses and is currently in phase I clinical development in HIV. We tested the capacity of two injections of the vaccine (10υg, i.p) given with or without poly(IC) (50υg, i.p) at 3 weeks apart to i) elicit human (hu) B-, and huT-cell responses in NSG mice reconstituted with a Human Immune System (HIS mice), ii) protect against SARS-CoV2 infection in the hCD40xK18hACE2 transgenic mice. Results: We performed AIM assays and intracellular staining on spleen cells of HIS mice stimulated with overlapping peptide pools spanning the sequences of vaccine antigens. We found that both non-adjuvanted and adjuvanted vaccine efficiently induced SARS-CoV2-specific Th1 huCD4+ and huCD8+ T cells in all vaccinees compared to mock animals. SARS-CoV2-specific huCD4+ T cells were polyfunctional. We confirmed the presence of RBD-specific huCD8+ T cells in the vaccinated animals using HLA-I tetramers. A significant proportion of the multimer+ huCD8+ T cells were Tscm (CD45RA+ CD62L+ CD95+) cells in both vaccinated groups. Besides, we detected significant amounts of spike-IgG+ switched huB cells in all vaccinees. In SARS-CoV2 challenge experiments, we further showed that both vaccination settings significantly protected animals with a survival rate of 100%. Conclusion: We demonstrate that the targeting of SARS-CoV-2 epitopes to CD40 induces significant B and T cells with a long-term memory phenotype in HIS mice and the ability of the vaccine to ensure complete protection against SARS-CoV2 infection.

11.
Egyptian Journal of Medical Human Genetics ; 23(1), 2022.
Article in English | EMBASE | ID: covidwho-1822226

ABSTRACT

Background: As the new pandemic created by COVID-19 virus created the need of rapid acquisition of a suitable vaccine against SARS-CoV-2 to develop Immunity and to reduce the mortality, the aim of this study was to identify SARS-CoV-2 S protein and N antigenic epitopes by using immunoinformatic methods to design a vaccine against SARS-CoV-2, for which S and N protein-dependent epitopes are predicted. B cell, CTL and HTL were determined based on antigenicity, allergenicity and toxicity that were non-allergenic, non-toxic, and antigenic and were selected for the design of a multi-epitope vaccine structure. Then, in order to increase the safety of Hbd-3 and Hbd-2 as adjuvants, they were connected to the N and C terminals of the vaccine construct, respectively, with a linker. The three-dimensional structure of the structure was predicted and optimized, and its quality was evaluated. The vaccine construct was ligated to MHCI. Finally, after optimizing the codon to increase expression in E. coli K12, the vaccine construct was cloned into pET28a (+) vector. Results: Epitopes which were used in our survey were based on non-allergenic, non-toxic and antigenic. Therefore, 543-amino-acid-long multi-epitope vaccine formation was invented through linking 9 cytotoxic CTL, 5 HTL and 14 B cell epitopes with appropriate adjuvants and connectors that can control the SARS coronavirus 2 infection and could be more assessed in medical scientific researches. Conclusion: We believe that the proposed multi-epitope vaccine can effectively evoke an immune response toward SARS-CoV-2.

12.
Biocell ; 46(SUPPL 1):180, 2022.
Article in English | EMBASE | ID: covidwho-1675776

ABSTRACT

The SARS-CoV-2 coronavirus, which causes respiratory syndrome COVID-19, has a protein nucleocapsid that envelops the viral ssRNA. The main protein of the nucleocapsid is the Np protein, which presents limited homology with nucleoproteins of other coronaviruses and therefore turns out to be an attractive antigen for the development of specific anti-Np antibodies. These antibodies can be used for the development of diagnostic systems that allow the detection of the viral antigen in infected individuals from saliva samples. In this context, our group has developed a labelling system called FasTAG®, which allows the immobilization of recombinant proteins on the surface of Gram+ formaldehyde inactivated bacteria. In this system, the recombinant proteins expressed in heterologous systems are fused to the C-terminal domain of S-Layer proteins of Lactobacillus sp. Then, the intrinsic affinity this domain possesses for the membranes of Gram+ bacteria is used for the immobilization of the recombinant proteins of interest. In this way, it is possible to purify specific antibodies against an antigen of interest. Based on the above, the objective of this work was to evaluate the functionality of the FasTAG® system to purify specific anti-Np antibodies. For this, the recombinant protein Np-FasTAG® was incubated for 12 hours at 4 °C with a matrix made up of B. subtilis inactivated with 3% formaldehyde. Next, for the optimization of the protein fixation process to the matrix, a compound factorial design was carried out, the variables of which were: formaldehyde concentration (0.5-1.5-2.5% v/v) and time of incubation (15-30-45 minutes). The optimal condition was determined as the one that minimizes the detachment of the Np protein and maximizes the detachment of the specific antibodies. Turning out to be the optimal condition for the elaboration of the affinity matrix 2.5% v/v of formaldehyde and 15 minutes. Then, in order to evaluate the application of the affinity matrix in the purification of specific antibodies, it was incubated for 1 hour with polyclonal antibodies obtained from chicken egg yolks and the serum of goats immunized with the Np antigen. Next, to study the elution conditions of the antibodies, a compound factorial design was performed using variables: pH, time, and SDS concentration. The best elution condition was obtained for pH 10.5 and 15 minutes. Subsequently, the purified antibodies were evaluated by SDS-PAGE and ELISA. As a result, it was possible to purify 3.5 μg of anti-Np IgG and 3.1 μg of anti-Np IgY per mg of resin. Finally, the set of experiments carried out here demonstrate the potential and functionality of this system for the purification of specific anti-Np antibodies and their use for diagnostic purposes.

SELECTION OF CITATIONS
SEARCH DETAIL