Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 518
Filter
1.
BMC Vet Res ; 20(1): 342, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095820

ABSTRACT

BACKGROUND: Porcine deltacoronavirus (PDCoV) is a swine enteropathogenic coronavirus that affects young pigs, causing vomiting, acute diarrhea, dehydration, and even death. There is growing evidence that PDCoV can undergo cross-species as well as zoonotic transmissions. Due to the frequent outbreaks of this deadly virus, early detection is essential for effective prevention and control. Therefore, developing a more convenient and reliable method for PDCoV detection is the need of the hour. RESULTS: This study utilized a high-affinity monoclonal antibody as the capture antibody and a horseradish peroxidase labeled polyclonal antibody as the detection antibody to develop an enzyme-linked immunosorbent assay (DAS-ELSA) for PDCoV detection.Both antibodies target the PDCoV nucleocapsid (N) protein. The findings of this study revealed that DAS-ELISA was highly specific to PDCoV and did not cross-react with other viruses to cause swine diarrhea. The limit of detection of the virus titer using this method was 103 TCID50/mL of PDCoV particles. The results of a parallel analysis of 239 known pig samples revealed a coincidence rate of 97.07% (κ = 0.922) using DAS-ELISA and reverse transcriptase PCR (RT-PCR). The DAS-ELISA was used to measure the one-step growth curve of PDCoV in LLC-PK cells and the tissue distribution of PDCoV in infected piglets. The study found that the DAS-ELISA was comparable in accuracy to the TCID50 method while measuring the one-step growth curve. Furthermore, the tissue distribution measured by DAS-ELISA was also consistent with the qRT-PCR method. CONCLUSION: The developed DAS-ELISA method can be conveniently used for the early clinical detection of PDCoV infection in pigs, and it may also serve as an alternative method for laboratory testing of PDCoV.


Subject(s)
Deltacoronavirus , Enzyme-Linked Immunosorbent Assay , Swine Diseases , Animals , Enzyme-Linked Immunosorbent Assay/veterinary , Enzyme-Linked Immunosorbent Assay/methods , Swine , Swine Diseases/virology , Swine Diseases/diagnosis , Swine Diseases/immunology , Deltacoronavirus/isolation & purification , Coronavirus Infections/veterinary , Coronavirus Infections/diagnosis , Coronavirus Infections/virology , Coronavirus Infections/immunology , Antibodies, Monoclonal/immunology , Sensitivity and Specificity , Antigens, Viral/analysis , Antigens, Viral/immunology , Antibodies, Viral/blood
2.
Methods Mol Biol ; 2824: 319-334, 2024.
Article in English | MEDLINE | ID: mdl-39039420

ABSTRACT

The nucleocapsid protein (N) in Rift Valley fever virus is an RNA-binding protein that functions in viral transcription, replication, and packaging. In this chapter, the method for studying protein-RNA interactions in context of viral infection using individual nucleotide resolution, cross-linking, immunoprecipitation, and sequencing (iCLIP-seq) is explained. The method is useful for identifying the interactions between both host and viral RNAs with N and can identify RNA motifs that interact with the protein of interest.


Subject(s)
Immunoprecipitation , Nucleocapsid Proteins , RNA, Viral , Rift Valley fever virus , Nucleocapsid Proteins/metabolism , RNA, Viral/metabolism , RNA, Viral/genetics , Binding Sites , Rift Valley fever virus/genetics , Rift Valley fever virus/metabolism , Immunoprecipitation/methods , Protein Binding , Humans , RNA-Binding Proteins/metabolism , High-Throughput Nucleotide Sequencing/methods
3.
Pathogens ; 13(7)2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39057809

ABSTRACT

INTRODUCTION: Rift Valley fever virus (RVFV) belonging to the Phenuiviridae family is responsible for a zoonotic disease called Rift Valley fever (RVF). Currently, RVFV has spread from Africa to Asia, and due to its ability to cause high mortality rates, it has significantly impacted human health and economic development in many societies. Highly specific and sensitive systems for sero-diagnosis of RVFV infection are needed for clinical use. METHOD: BALB/c mice were immunized with recombinant RVFV nucleocapsid (rRVFV-N) protein and the spleen cells fused with SP2/0 myeloma cells to create hybridoma cell lines. The secreted monoclonal antibodies (MAbs) were purified and characterized. Enzyme-linked immunosorbent assay (ELISA) systems for the detection of IgG and IgM using the new MAbs were established and evaluated. Serum samples from 96 volunteers and 93 patients of suspected RVF from Kenya were tested compared with the ELISA systems based on inactivated viruses and the rabbit polyclonal antibody. RESULT: Three monoclonal antibodies against rRVFV-N protein were established. The performance of the MAb-based sandwich IgG ELISA and the IgM capture ELISA perfectly matched the ELISA systems using the inactivated virus or the polyclonal antibody. CONCLUSIONS: Recombinant RVFV-N protein-specific MAbs were developed and they offer useful tools for RVFV studies. The MAb-based ELISA systems for detecting IgG and IgM offer safe and useful options for diagnosing RVFV infections in humans.

4.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000276

ABSTRACT

Neurologic manifestations are an immediate consequence of SARS-CoV-2 infection, the etiologic agent of COVID-19, which, however, may also trigger long-term neurological effects. Notably, COVID-19 patients with neurological symptoms show elevated levels of biomarkers associated with brain injury, including Tau proteins linked to Alzheimer's pathology. Studies in brain organoids revealed that SARS-CoV-2 alters the phosphorylation and distribution of Tau in infected neurons, but the mechanisms are currently unknown. We hypothesize that these pathological changes are due to the recruitment of Tau into stress granules (SGs) operated by the nucleocapsid protein (NCAP) of SARS-CoV-2. To test this hypothesis, we investigated whether NCAP interacts with Tau and localizes to SGs in hippocampal neurons in vitro and in vivo. Mechanistically, we tested whether SUMOylation, a posttranslational modification of NCAP and Tau, modulates their distribution in SGs and their pathological interaction. We found that NCAP and Tau colocalize and physically interact. We also found that NCAP induces hyperphosphorylation of Tau and causes cognitive impairment in mice infected with NCAP in their hippocampus. Finally, we found that SUMOylation modulates NCAP SG formation in vitro and cognitive performance in infected mice. Our data demonstrate that NCAP induces Tau pathological changes both in vitro and in vivo. Moreover, we demonstrate that SUMO2 ameliorates NCAP-induced Tau pathology, highlighting the importance of the SUMOylation pathway as a target of intervention against neurotoxic insults, such as Tau oligomers and viral infection.


Subject(s)
COVID-19 , Coronavirus Nucleocapsid Proteins , Hippocampus , Neurons , SARS-CoV-2 , Sumoylation , tau Proteins , tau Proteins/metabolism , Animals , Mice , Humans , Hippocampus/metabolism , Hippocampus/pathology , COVID-19/metabolism , COVID-19/pathology , COVID-19/virology , SARS-CoV-2/pathogenicity , SARS-CoV-2/metabolism , Phosphorylation , Coronavirus Nucleocapsid Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Neurons/virology , Small Ubiquitin-Related Modifier Proteins/metabolism , Stress Granules/metabolism , Mice, Inbred C57BL , Phosphoproteins/metabolism , Male , Nucleocapsid Proteins/metabolism , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/virology
5.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39000451

ABSTRACT

The nucleocapsid (N) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a viral structural protein that is abundant in the circulation of infected individuals. Previous published studies reported controversial data about the role of the N protein in the activation of the complement system. It was suggested that the N protein directly interacts with mannose-binding lectin-associated serine protease-2 (MASP-2) and stimulates lectin pathway overactivation/activity. In order to check these data and to reveal the mechanism of activation, we examined the effect of the N protein on lectin pathway activation. We found that the N protein does not bind to MASP-2 and MASP-1 and it does not stimulate lectin pathway activity in normal human serum. Furthermore, the N protein does not facilitate the activation of zymogen MASP-2, which is MASP-1 dependent. Moreover, the N protein does not boost the enzymatic activity of MASP-2 either on synthetic or on protein substrates. In some of our experiments, we observed that MASP-2 digests the N protein. However, it is questionable, whether this activity is biologically relevant. Although surface-bound N protein did not activate the lectin pathway, it did trigger the alternative pathway in 10% human serum. Additionally, we detected some classical pathway activation by the N protein. Nevertheless, we demonstrated that this activation was induced by the bound nucleic acid, rather than by the N protein itself.


Subject(s)
Complement Pathway, Mannose-Binding Lectin , Coronavirus Nucleocapsid Proteins , Mannose-Binding Protein-Associated Serine Proteases , SARS-CoV-2 , Humans , Mannose-Binding Protein-Associated Serine Proteases/metabolism , SARS-CoV-2/metabolism , Coronavirus Nucleocapsid Proteins/metabolism , COVID-19/virology , COVID-19/metabolism , COVID-19/immunology , Phosphoproteins/metabolism , Protein Binding , Complement Activation
6.
J Virol Methods ; 329: 114995, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38972641

ABSTRACT

Diagnostics employing multiple modalities have been essential for controlling and managing COVID-19, caused by SARS-CoV-2. However, scaling up Reverse Transcription-Quantitative Polymerase Chain Reaction (RT-qPCR), the gold standard for SARS-CoV-2 detection, remains challenging in low and middle-income countries. Cost-effective and high-throughput alternatives like enzyme-linked immunosorbent assay (ELISA) could address this issue. We developed an in-house SARS-CoV-2 nucleocapsid capture ELISA, and validated on 271 nasopharyngeal swab samples from humans (n = 252), bovines (n = 10), and dogs (n = 9). This ELISA has a detection limit of 195 pg/100 µL of nucleocapsid protein and does not cross-react with related coronaviruses, ensuring high specificity to SARS-CoV-2. Diagnostic performance was evaluated using receiver operating characteristic curve analysis, showing a diagnostic sensitivity of 67.78 % and specificity of 100 %. Sensitivity improved to 74.32 % when excluding positive clinical samples with RT-qPCR Ct values > 25. Furthermore, inter-rater reliability analysis demonstrated substantial agreement (κ values = 0.73-0.80) with the VIRALDTECT II Multiplex RT-qPCR kit and perfect agreement with the CoVeasy™ COVID-19 rapid antigen self-test (κ values = 0.89-0.93). Our findings demonstrated that the in-house nucleocapsid capture ELISA is suitable for SARS-CoV-2 testing in humans and animals, meeting the necessary sensitivity and specificity thresholds for cost-effective, large-scale screening.

7.
Virology ; 597: 110163, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38959724

ABSTRACT

To gain insight into the functional relationship between the nucleocapsid (NC) domains of the Gag polyproteins of feline and simian immunodeficiency viruses, FIV and SIV, respectively, we generated two FIV Gag chimeric proteins containing different SIV NC and gag sequences. A chimeric FIV Gag protein (NC1) containing the SIV two zinc fingers motifs was incapable of assembling into virus-like particles. By contrast, another Gag chimera (NC2) differing from NC1 by the replacement of the C-terminal region of the FIV NC with SIV SP2 produced particles as efficiently as wild-type FIV Gag. Of note, when the chimeric NC2 Gag polyprotein was expressed in the context of the proviral DNA in feline CrFK cells, wild-type levels of virions were produced which encapsidated 50% of genomic RNA when compared to the wild-type virus.


Subject(s)
Gene Products, gag , Immunodeficiency Virus, Feline , Simian Immunodeficiency Virus , Virus Assembly , Zinc Fingers , Animals , Immunodeficiency Virus, Feline/genetics , Immunodeficiency Virus, Feline/metabolism , Immunodeficiency Virus, Feline/physiology , Gene Products, gag/genetics , Gene Products, gag/metabolism , Gene Products, gag/chemistry , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , Cats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Cell Line , Nucleocapsid/metabolism , Nucleocapsid/genetics , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Phenotype
8.
Biomolecules ; 14(7)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-39062463

ABSTRACT

Highly pathogenic coronaviruses have caused significant outbreaks in humans and animals, posing a serious threat to public health. The rapid global spread of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has resulted in millions of infections and deaths. However, the mechanisms through which coronaviruses evade a host's antiviral immune system are not well understood. Liquid-liquid phase separation (LLPS) is a recently discovered mechanism that can selectively isolate cellular components to regulate biological processes, including host antiviral innate immune signal transduction pathways. This review focuses on the mechanism of coronavirus-induced LLPS and strategies for utilizing LLPS to evade the host antiviral innate immune response, along with potential antiviral therapeutic drugs and methods. It aims to provide a more comprehensive understanding and novel insights for researchers studying LLPS induced by pandemic viruses.


Subject(s)
COVID-19 , Immunity, Innate , Animals , Humans , COVID-19/virology , Phase Separation , Signal Transduction
9.
Cell Rep Methods ; 4(7): 100818, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38986614

ABSTRACT

Protein-protein interactions play an important biological role in every aspect of cellular homeostasis and functioning. Proximity labeling mass spectrometry-based proteomics overcomes challenges typically associated with other methods and has quickly become the current state of the art in the field. Nevertheless, tight control of proximity-labeling enzymatic activity and expression levels is crucial to accurately identify protein interactors. Here, we leverage a T2A self-cleaving peptide and a non-cleaving mutant to accommodate the protein of interest in the experimental and control TurboID setup. To allow easy and streamlined plasmid assembly, we built a Golden Gate modular cloning system to generate plasmids for transient expression and stable integration. To highlight our T2A Split/link design, we applied it to identify protein interactions of the glucocorticoid receptor and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleocapsid and non-structural protein 7 (NSP7) proteins by TurboID proximity labeling. Our results demonstrate that our T2A split/link provides an opportune control that builds upon previously established control requirements in the field.


Subject(s)
Peptides , Proteomics , SARS-CoV-2 , Proteomics/methods , Humans , SARS-CoV-2/metabolism , SARS-CoV-2/genetics , Peptides/metabolism , Peptides/chemistry , COVID-19/metabolism , COVID-19/virology , HEK293 Cells , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/chemistry , Coronavirus Nucleocapsid Proteins/metabolism , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/chemistry , Plasmids/genetics , Plasmids/metabolism , Mass Spectrometry/methods , Phosphoproteins/metabolism , Phosphoproteins/genetics , Protein Interaction Mapping/methods
10.
J Virol ; : e0092624, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39082816

ABSTRACT

The swine acute diarrhea syndrome coronavirus (SADS-CoV) has caused significant disruptions in porcine breeding and raised concerns about potential human infection. The nucleocapsid (N) protein of SADS-CoV plays a vital role in viral assembly and replication, but its structure and functions remain poorly understood. This study utilized biochemistry, X-ray crystallography, and immunization techniques to investigate the N protein's structure and function in SADS-CoV. Our findings revealed distinct domains within the N protein, including an RNA-binding domain, two disordered domains, and a dimerization domain. Through biochemical assays, we confirmed that the N-terminal domain functions as an RNA-binding domain, and the C-terminal domain is involved in dimerization, with the crystal structure analysis providing visual evidence of dimer formation. Immunization experiments demonstrated that the disordered domain 2 elicited a significant antibody response. These identified domains and their interactions are crucial for viral assembly. This comprehensive understanding of the N protein in SADS-CoV enhances our knowledge of its assembly and replication mechanisms, enabling the development of targeted interventions and therapeutic strategies. IMPORTANCE: SADS-CoV is a porcine coronavirus that originated from a bat HKU2-related coronavirus. It causes devastating swine diseases and poses a high risk of spillover to humans. The coronavirus N protein, as the most abundant viral protein in infected cells, likely plays a key role in viral assembly and replication. However, the structure and function of this protein remain unclear. Therefore, this study employed a combination of biochemistry and X-ray crystallography to uncover distinct structural domains in the N protein, including RNA-binding domains, two disordered domains, and dimerization domains. Additionally, we made the novel discovery that the disordered domain elicited a significant antibody response. These findings provide new insights into the structure and functions of the SADS-CoV N protein, which have important implications for future studies on SADS-CoV diagnosis, as well as the development of vaccines and anti-viral drugs.

11.
Vaccine X ; 19: 100518, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39040888

ABSTRACT

Background: Our investigation focused whether infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) before or after receiving the mRNA COVID-19 vaccine can increase immune protection. And we also investigated relationship of infection acquired. Methods: Three shots of the mRNA coronavirus disease 2019 (COVID-19) vaccine BNT162b2 were administered to 736 healthcare workers at Tokyo Shinagawa Hospital. Serum samples were collected before the first shot (P1), at one month (P2), and at six months (P3) after the second shot and at one month after the third shot (P4). The presence of infection was assessed using IgG against the nucleocapsid (IgG (N) and RBD in the spike protein of SARS-CoV-2. We defined infection before P2 as natural infection (NI) and infection between P2 and P3 as breakthrough infection (BI) and compared susceptibility to further infection between the NI (-) and NI (+) groups and between BI (-) and BI (+) groups. Events in 485 participants who had a complete dataset of IgG (N) and IgG (RBD) from P1 to P4 were analyzed. Results: The presence of SARS-CoV-2 infection before P2 were examined by examining the titers of IgG (N)P1, IgG (N) P2, and IgG (RBD) P1 that exceeded the cutoff values. Consequently, 35 participants (7.22 %) were categorized into the NI (+) group, whereas 450 (92.8 %) were categorized into the NI (-) group. Between P2 and P3, the NI (-) group showed a higher rate of SARS-CoV-2 infection than the NI (+) group; however, there was no significant difference in the infection rate between P3 and P4. The infection rate was significantly lower in the BI (+) group than in the BI (-) group. Pre-primary vaccination infection significantly increased IgG (RBD) levels between P1 and P3. Post-primary vaccination infection significantly increased IgG (RBD) levels between P3 and P4. Conclusions: Infection with SARS-CoV-2 before or after receiving the mRNA COVID-19 vaccine can increase immune protection; however, the duration of this effect may be limited.

12.
Front Immunol ; 15: 1418678, 2024.
Article in English | MEDLINE | ID: mdl-39021574

ABSTRACT

Background: Knowledge about SARS-CoV-2 antibody dynamics in neonates and direct comparisons with maternal antibody responses are not well established. This study aimed to characterize and directly compare the maternal and infant antibody response in a national birth cohort from the Faroe Islands. Methods: The levels of immunoglobulins (Ig) targeting the receptor binding domain (RBD) of the spike protein and the nucleocapsid protein (N protein) of SARS-CoV-2 were investigated in maternal blood and umbilical cord blood from neonates. The study included 537 neonates and 565 mothers from the Faroe Islands, and follow-up samples were collected 12 months after birth. Multiple linear regression models were used to assess associations of maternal parameters with maternal and neonatal Ig levels and pregnancy outcomes. Results: The finding showed that neonates acquired varying levels of SARS-CoV-2 antibodies through transplacental transfer, and the levels were significantly influenced by the mother's vaccination and infection status. The study also found that maternal vaccination and the presence of SARS-CoV-2 antibodies targeting spike RBD were associated with gestational age and APGAR scores. Furthermore, the anti-RBD and -N protein-specific antibody response dynamics during 12 months after birth exhibited differences between mothers and children. RBD and N protein responses were maintained at follow-up in the mother's cohort, while only the N protein response was maintained at follow-up in the children's cohort. Conclusion: In conclusion, SARS-CoV-2-specific immune responses in newborns rely on maternal immunity, while the persistence of SARS-CoV-2-specific Igs appears to be differently regulated between mothers and children. The study provides new insights into the dynamics of SARS-CoV-2-specific immune responses in newborns and underscores the nuanced relationship between maternal factors and neonatal humoral responses.


Subject(s)
Antibodies, Viral , COVID-19 , Coronavirus Nucleocapsid Proteins , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , Female , SARS-CoV-2/immunology , COVID-19/immunology , Pregnancy , Antibodies, Viral/blood , Antibodies, Viral/immunology , Spike Glycoprotein, Coronavirus/immunology , Infant, Newborn , Coronavirus Nucleocapsid Proteins/immunology , Adult , Immunity, Maternally-Acquired , Infant , Male , Cohort Studies , Phosphoproteins/immunology , Pregnancy Complications, Infectious/immunology , Fetal Blood/immunology
13.
Viruses ; 16(7)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39066193

ABSTRACT

Puumala orthohantavirus (PUUV) is an emerging zoonotic virus endemic to Europe and Russia that causes nephropathia epidemica, a mild form of hemorrhagic fever with renal syndrome (HFRS). There are limited options for treatment and diagnosis of orthohantavirus infection, making the search for potential immunogenic candidates crucial. In the present work, various bioinformatics tools were employed to design conserved immunogenic peptides containing multiple epitopes of PUUV nucleocapsid protein. Eleven conserved peptides (90% conservancy) of the PUUV nucleocapsid protein were identified. Three conserved peptides containing multiple T and B cell epitopes were selected using a consensus epitope prediction algorithm. Molecular docking using the HPEP dock server demonstrated strong binding interactions between the epitopes and HLA molecules (ten alleles for each class I and II HLA). Moreover, an analysis of population coverage using the IEDB database revealed that the identified peptides have over 90% average population coverage across six continents. Molecular docking and simulation analysis reveal a stable interaction with peptide constructs of chosen immunogenic peptides and Toll-like receptor-4. These computational analyses demonstrate selected peptides' immunogenic potential, which needs to be validated in different experimental systems.


Subject(s)
Molecular Docking Simulation , Nucleocapsid Proteins , Peptides , Puumala virus , Puumala virus/immunology , Puumala virus/genetics , Peptides/immunology , Peptides/chemistry , Humans , Nucleocapsid Proteins/immunology , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Hemorrhagic Fever with Renal Syndrome/immunology , Hemorrhagic Fever with Renal Syndrome/virology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/chemistry , Computational Biology , Conserved Sequence , Amino Acid Sequence , Protein Binding
14.
Viruses ; 16(7)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39066279

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of COVID-19 and responsible for the global coronavirus pandemic which started in 2019. Despite exhaustive efforts to trace its origins, including potential links with pangolins and bats, the precise origins of the virus remain unclear. Bats have been recognized as natural hosts for various coronaviruses, including the Middle East respiratory coronavirus (MERS-CoV) and the SARS-CoV. This study presents a comparative analysis of the SARS-CoV-2 nucleocapsid protein (N) interactome in human and bat cell lines. We identified approximately 168 cellular proteins as interacting partners of SARS-CoV-2 N in human cells and 196 cellular proteins as interacting partners with this protein in bat cells. The results highlight pathways and events that are both common and unique to either bat or human cells. Understanding these interactions is crucial to comprehend the reasons behind the remarkable resilience of bats to viral infections. This study provides a foundation for a deeper understanding of host-virus interactions in different reservoirs.


Subject(s)
COVID-19 , Chiroptera , Coronavirus Nucleocapsid Proteins , Phosphoproteins , Proteomics , SARS-CoV-2 , Chiroptera/virology , Humans , SARS-CoV-2/metabolism , Animals , Coronavirus Nucleocapsid Proteins/metabolism , Cell Line , Proteomics/methods , Phosphoproteins/metabolism , COVID-19/virology , COVID-19/metabolism , Host-Pathogen Interactions , Protein Interaction Maps
15.
Future Sci OA ; 10(1): FSO917, 2024.
Article in English | MEDLINE | ID: mdl-38827795

ABSTRACT

Aims: To investigate the role of phosphorylation in SARS-CoV-2 infection, potential therapeutic targets and its harmful genetic sequences. Materials & Methods: Data mining techniques were employed to identify upregulated kinases responsible for proteomic changes induced by SARS-CoV-2. Spike and nucleocapsid proteins' sequences were analyzed using predictive tools, including SNAP2, MutPred2, PhD-SNP, SNPs&Go, MetaSNP, Predict-SNP and PolyPhen-2. Missense variants were identified using ensemble-based algorithms and homology/structure-based models like SIFT, PROVEAN, Predict-SNP and MutPred-2. Results: Eight missense variants were identified in viral sequences. Four damaging variants were found, with SNPs&Go and PolyPhen-2. Promising therapeutic candidates, including gilteritinib, pictilisib, sorafenib, RO5126766 and omipalisib, were identified. Conclusion: This research offers insights into SARS-CoV-2 pathogenicity, highlighting potential treatments and harmful variants in viral proteins.


This study explores the process called phosphorylation, which involves adding phosphate groups to certain proteins, influences the way the SARS-CoV-2 virus causes disease. The virus manipulates host enzymes to help it spread and survive. Researchers used data analysis techniques to identify the proteins that play a role in this process, aiming to find potential targets for treatments. They analyzed genetic sequences of key virus proteins and used various tools to predict harmful mutations. The study found several promising compounds that could be used to target the virus. Further research and experiments are needed to confirm their effectiveness as COVID-19 treatments.


This research explored the process called phosphorylation, which involves adding certain molecules to proteins, affects how the SARS-CoV-2 virus makes people sick. The virus uses our own cell's machinery to help it spread. Researchers used computer analysis to find out which proteins are involved in this process, hoping to find new ways to treat COVID-19. They studied the genetic code of important parts of the virus and used computer programs to predict if there were harmful changes in the code. They found some potential medicines that could be used to fight the virus and reduce its harm, but more research and testing are needed to be sure.

16.
Acta Pharm Sin B ; 14(6): 2505-2519, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38828154

ABSTRACT

The nucleocapsid protein (NP) plays a crucial role in SARS-CoV-2 replication and is the most abundant structural protein with a long half-life. Despite its vital role in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) assembly and host inflammatory response, it remains an unexplored target for drug development. In this study, we identified a small-molecule compound (ciclopirox) that promotes NP degradation using an FDA-approved library and a drug-screening cell model. Ciclopirox significantly inhibited SARS-CoV-2 replication both in vitro and in vivo by inducing NP degradation. Ciclopirox induced abnormal NP aggregation through indirect interaction, leading to the formation of condensates with higher viscosity and lower mobility. These condensates were subsequently degraded via the autophagy-lysosomal pathway, ultimately resulting in a shortened NP half-life and reduced NP expression. Our results suggest that NP is a potential drug target, and that ciclopirox holds substantial promise for further development to combat SARS-CoV-2 replication.

17.
Front Cell Infect Microbiol ; 14: 1415885, 2024.
Article in English | MEDLINE | ID: mdl-38846351

ABSTRACT

Corona Virus Disease 2019 (COVID-19) is a highly prevalent and potent infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Until now, the world is still endeavoring to develop new ways to diagnose and treat COVID-19. At present, the clinical prevention and treatment of COVID-19 mainly targets the spike protein on the surface of SRAS-CoV-2. However, with the continuous emergence of SARS-CoV-2 Variants of concern (VOC), targeting the spike protein therapy shows a high degree of limitation. The Nucleocapsid Protein (N protein) of SARS-CoV-2 is highly conserved in virus evolution and is involved in the key process of viral infection and assembly. It is the most expressed viral structural protein after SARS-CoV-2 infection in humans and has high immunogenicity. Therefore, N protein as the key factor of virus infection and replication in basic research and clinical application has great potential research value. This article reviews the research progress on the structure and biological function of SARS-CoV-2 N protein, the diagnosis and drug research of targeting N protein, in order to promote researchers' further understanding of SARS-CoV-2 N protein, and lay a theoretical foundation for the possible outbreak of new and sudden coronavirus infectious diseases in the future.


Subject(s)
COVID-19 , Coronavirus Nucleocapsid Proteins , Phosphoproteins , SARS-CoV-2 , SARS-CoV-2/genetics , Humans , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/metabolism , COVID-19/virology , COVID-19/diagnosis , Phosphoproteins/metabolism , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/genetics
18.
Viruses ; 16(6)2024 May 29.
Article in English | MEDLINE | ID: mdl-38932164

ABSTRACT

The HIV-1 nucleocapsid protein (NC) is a multifunctional viral protein necessary for HIV-1 replication. Recent studies have demonstrated that reverse transcription (RT) completes in the intact viral capsid, and the timing of RT and uncoating are correlated. How the small viral core stably contains the ~10 kbp double stranded (ds) DNA product of RT, and the role of NC in this process, are not well understood. We showed previously that NC binds and saturates dsDNA in a non-specific electrostatic binding mode that triggers uniform DNA self-attraction, condensing dsDNA into a tight globule against extending forces up to 10 pN. In this study, we use optical tweezers and atomic force microscopy to characterize the role of NC's basic residues in dsDNA condensation. Basic residue mutations of NC lead to defective interaction with the dsDNA substrate, with the constant force plateau condensation observed with wild-type (WT) NC missing or diminished. These results suggest that NC's high positive charge is essential to its dsDNA condensing activity, and electrostatic interactions involving NC's basic residues are responsible in large part for the conformation, size, and stability of the dsDNA-protein complex inside the viral core. We observe DNA re-solubilization and charge reversal in the presence of excess NC, consistent with the electrostatic nature of NC-induced DNA condensation. Previous studies of HIV-1 replication in the presence of the same cationic residue mutations in NC showed significant defects in both single- and multiple-round viral infectivity. Although NC participates in many stages of viral replication, our results are consistent with the hypothesis that cationic residue mutations inhibit genomic DNA condensation, resulting in increased premature capsid uncoating and contributing to viral replication defects.


Subject(s)
DNA, Viral , HIV-1 , Reverse Transcription , HIV-1/genetics , HIV-1/physiology , HIV-1/chemistry , HIV-1/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/chemistry , Humans , Cations/metabolism , Virus Replication , Microscopy, Atomic Force , Virion/metabolism , Virion/genetics , Virion/chemistry , Mutation
19.
Sensors (Basel) ; 24(12)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38931556

ABSTRACT

This paper reports a rapid and sensitive sensor for the detection and quantification of the COVID-19 N-protein (N-PROT) via an electrochemical mechanism. Single-frequency electrochemical impedance spectroscopy was used as a transduction method for real-time measurement of the N-PROT in an immunosensor system based on gold-conjugate-modified carbon screen-printed electrodes (Cov-Ag-SPE). The system presents high selectivity attained through an optimal stimulation signal composed of a 0.0 V DC potential and 10 mV RMS-1 AC signal at 100 Hz over 300 s. The Cov-Ag-SPE showed a log response toward N-PROT detection at concentrations from 1.0 ng mL-1 to 10.0 µg mL-1, with a 0.977 correlation coefficient for the phase (θ) variation. An ML-based approach could be created using some aspects observed from the positive and negative samples; hence, it was possible to classify 252 samples, reaching 83.0, 96.2 and 91.3% sensitivity, specificity, and accuracy, respectively, with confidence intervals (CI) ranging from 73.0 to 100.0%. Because impedance spectroscopy measurements can be performed with low-cost portable instruments, the immunosensor proposed here can be applied in point-of-care diagnostics for mass testing, even in places with limited resources, as an alternative to the common diagnostics methods.


Subject(s)
Biosensing Techniques , COVID-19 , Dielectric Spectroscopy , Gold , SARS-CoV-2 , COVID-19/diagnosis , COVID-19/virology , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , Humans , SARS-CoV-2/isolation & purification , SARS-CoV-2/immunology , Dielectric Spectroscopy/instrumentation , Dielectric Spectroscopy/methods , Gold/chemistry , Electrodes , Electrochemical Techniques/methods , Electrochemical Techniques/instrumentation , Immunoassay/methods , Immunoassay/instrumentation , Coronavirus Nucleocapsid Proteins/immunology , Coronavirus Nucleocapsid Proteins/analysis , Carbon/chemistry , Phosphoproteins/analysis
20.
J Virol ; 98(7): e0033424, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38829137

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an enteric pathogenic coronavirus that causes acute and severe watery diarrhea in piglets and has the ability of cross-species transmission, posing a great threat to swine production and public health. The interferon (IFN)-mediated signal transduction represents an important component of virus-host interactions and plays an essential role in regulating viral infection. Previous studies have suggested that multifunctional viral proteins encoded by coronaviruses antagonize the production of IFN via various means. However, the function of these viral proteins in regulating IFN-mediated signaling pathways is largely unknown. In this study, we demonstrated that PDCoV and its encoded nucleocapsid (N) protein antagonize type I IFN-mediated JAK-STAT signaling pathway. We identified that PDCoV infection stimulated but delayed the production of IFN-stimulated genes (ISGs). In addition, PDCoV inhibited JAK-STAT signal transduction by targeting the nuclear translocation of STAT1 and ISGF3 formation. Further evidence showed that PDCoV N is the essential protein involved in the inhibition of type I IFN signaling by targeting STAT1 nuclear translocation via its C-terminal domain. Mechanistically, PDCoV N targets STAT1 by interacting with it and subsequently inhibiting its nuclear translocation. Furthermore, PDCoV N inhibits STAT1 nuclear translocation by specifically targeting KPNA2 degradation through the lysosomal pathway, thereby inhibiting the activation of downstream sensors in the JAK-STAT signaling pathway. Taken together, our results reveal a novel mechanism by which PDCoV N interferes with the host antiviral response.IMPORTANCEPorcine deltacoronavirus (PDCoV) is a novel enteropathogenic coronavirus that receives increased attention and seriously threatens the pig industry and public health. Understanding the underlying mechanism of PDCoV evading the host defense during infection is essential for developing targeted drugs and effective vaccines against PDCoV. This study demonstrated that PDCoV and its encoded nucleocapsid (N) protein antagonize type I interferon signaling by targeting STAT1, which is a crucial signal sensor in the JAK-STAT signaling pathway. Further experiments suggested that PDCoV N-mediated inhibition of the STAT1 nuclear translocation involves the degradation of KPNA2, and the lysosome plays a role in KPNA2 degradation. This study provides new insights into the regulation of PDCoV N in the JAK-STAT signaling pathway and reveals a novel mechanism by which PDCoV evades the host antiviral response. The novel findings may guide us to discover new therapeutic targets and develop live attenuated vaccines for PDCoV infection.


Subject(s)
Deltacoronavirus , Nucleocapsid Proteins , STAT1 Transcription Factor , Signal Transduction , Animals , Swine , STAT1 Transcription Factor/metabolism , Deltacoronavirus/metabolism , Nucleocapsid Proteins/metabolism , Humans , Janus Kinases/metabolism , Swine Diseases/virology , Swine Diseases/metabolism , alpha Karyopherins/metabolism , Interferon Type I/metabolism , Coronavirus Infections/virology , Coronavirus Infections/metabolism , HEK293 Cells , Cell Line , Proteolysis , Host-Pathogen Interactions
SELECTION OF CITATIONS
SEARCH DETAIL