Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
PLoS Pathog ; 19(8): e1011532, 2023 08.
Article in English | MEDLINE | ID: mdl-37531329

ABSTRACT

The COVID-19 pandemic represents a global challenge that has impacted and is expected to continue to impact the lives and health of people across the world for the foreseeable future. The rollout of vaccines has provided highly anticipated relief, but effective therapeutics are required to further reduce the risk and severity of infections. Monoclonal antibodies have been shown to be effective as therapeutics for SARS-CoV-2, but as new variants of concern (VoC) continue to emerge, their utility and use have waned due to limited or no efficacy against these variants. Furthermore, cumbersome systemic administration limits easy and broad access to such drugs. As well, concentrations of systemically administered antibodies in the mucosal epithelium, a primary site of initial infection, are dependent on neonatal Fc receptor mediated transport and require high drug concentrations. To reduce the viral load more effectively in the lung, we developed an inhalable formulation of a SARS-CoV-2 neutralizing antibody binding to a conserved epitope on the Spike protein, ensuring pan-neutralizing properties. Administration of this antibody via a vibrating mesh nebulization device retained antibody integrity and resulted in effective distribution of the antibody in the upper and lower respiratory tract of non-human primates (NHP). In comparison with intravenous administration, significantly higher antibody concentrations can be obtained in the lung, resulting in highly effective reduction in viral load post SARS-CoV-2 challenge. This approach may reduce the barriers of access and uptake of antibody therapeutics in real-world clinical settings and provide a more effective blueprint for targeting existing and potentially emerging respiratory tract viruses.


Subject(s)
Antiviral Agents , COVID-19 , Animals , Humans , SARS-CoV-2 , Pandemics , Antibodies, Viral , Antibodies, Neutralizing , Epitopes , Spike Glycoprotein, Coronavirus
2.
Cell Rep Med ; 3(8): 100716, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35952669

ABSTRACT

The high number of mutations in the Omicron variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes its immune escape. We report a longitudinal analysis of 111 vaccinated individuals for their antibody levels up to 6 months after the third dose of the BNT162b2 vaccine. After the third dose, the antibody levels decline but less than after the second dose. The booster dose remarkably increases the serum ability to block wild-type or Omicron variant spike protein's receptor-binding domain (RBD) interaction with the angiotensin-converting enzyme 2 (ACE2) receptor, and these protective antibodies persist 3 months later. Three months after the booster dose, memory CD4+ and CD8+ T cells to the wild-type and Omicron variant are detectable in the majority of vaccinated individuals. Our data show that the third dose restores the high levels of blocking antibodies and enhances T cell responses to Omicron.


Subject(s)
COVID-19 , Vaccines , Antibodies , BNT162 Vaccine , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , COVID-19/prevention & control , Humans , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Viral Envelope Proteins/chemistry
3.
PLoS One ; 17(6): e0268806, 2022.
Article in English | MEDLINE | ID: mdl-35687549

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to impose a serious burden on health systems globally. Despite worldwide vaccination, social distancing and wearing masks, the spread of the virus is ongoing. One of the mechanisms by which neutralizing antibodies (NAbs) block virus entry into cells encompasses interaction inhibition between the cell surface receptor angiotensin-converting enzyme 2 (ACE2) and the spike (S) protein of SARS-CoV-2. SARS-CoV-2-specific NAb development can be induced in the blood of cattle. Pregnant cows produce NAbs upon immunization, and antibodies move into the colostrum immediately before calving. Here, we immunized cows with SARS-CoV-2 S1 receptor binding domain (RBD) protein in proper adjuvant solutions, followed by one boost with SARS-CoV-2 trimeric S protein and purified immunoglobulins from colostrum. We demonstrate that this preparation indeed blocks the interaction between the trimeric S protein and ACE2 in different in vitro assays. Moreover, we describe the formulation of purified immunoglobulin preparation into a nasal spray. When administered to human subjects, the formulation persisted on the nasal mucosa for at least 4 hours, as determined by a clinical study. Therefore, we are presenting a solution that shows great potential to serve as a prophylactic agent against SARS-CoV-2 infection as an additional measure to vaccination and wearing masks. Moreover, our technology allows for rapid and versatile adaptation for preparing prophylactic treatments against other diseases using the defined characteristics of antibody movement into the colostrum.


Subject(s)
COVID-19 , SARS-CoV-2 , Angiotensin-Converting Enzyme 2 , Animals , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , Cattle , Colostrum/metabolism , Female , Humans , Pregnancy , Spike Glycoprotein, Coronavirus
4.
J Virol ; 94(20)2020 09 29.
Article in English | MEDLINE | ID: mdl-32759324

ABSTRACT

The life cycle of human papillomaviruses (HPVs) comprises three distinct phases of DNA replication: initial amplification, maintenance of the genome copy number at a constant level, and vegetative amplification. The viral helicase E1 is one of the factors required for the initiation of HPV genome replication. However, the functions of the E1 protein during other phases of the viral life cycle are largely uncharacterized. Here, we studied the role of the HPV18 E1 helicase in three phases of viral genome replication by downregulating E1 expression using RNA interference or inducing degradation of the E1 protein via inhibition of casein kinase 2α expression or catalytic activity. We generated a novel modified HPV18 genome expressing Nanoluc and tagged E1 and E2 proteins and created several stable HPV18-positive cell lines. We showed that, in contrast to initial amplification of the HPV18 genome, other phases of viral genome replication involve also an E1-independent mechanism. We characterize two distinct populations of HPV18 replicons existing during the maintenance and vegetative amplification phases. We show that a subset of these replicons, including viral genome monomers, replicate in an E1-dependent manner, while some oligomeric forms of the HPV18 genome replicate independently of E1 function.IMPORTANCE Human papillomavirus (HPV) infections pose serious medical problem. To date, there are no HPV-specific antivirals available due to poor understanding of the molecular mechanisms of virus infection cycle. The infection cycle of HPV involves initial amplification of the viral genomes and maintenance of the viral genomes with a constant copy number, followed by another round of viral genome amplification and new viral particle formation. The viral protein E1 is critical for the initial amplification of the viral genome. However, E1 involvement in other phases of the viral life cycle has remained controversial. In the present study, we show that at least two different replication modes of the HPV18 genome are undertaken simultaneously during the maintenance and vegetative amplification phases, i.e., replication of the majority of the HPV18 genome proceeds under the control of the host cell replication machinery without E1 function, whereas a minority of the genome replicates in an E1-dependent manner.


Subject(s)
Gene Expression Regulation, Viral , Genome, Viral , Human papillomavirus 18/physiology , Oncogene Proteins, Viral/metabolism , Virus Replication , Cell Line, Tumor , Humans , Oncogene Proteins, Viral/genetics
5.
PLoS Pathog ; 15(5): e1007788, 2019 05.
Article in English | MEDLINE | ID: mdl-31091289

ABSTRACT

Inhibition of human papillomavirus (HPV) replication is a promising therapeutic approach for intervening with HPV-related pathologies. Primary targets for interference are two viral proteins, E1 and E2, which are required for HPV replication. Both E1 and E2 are phosphoproteins; thus, the protein kinases that phosphorylate them might represent secondary targets to achieve inhibition of HPV replication. In the present study, we show that CX4945, an ATP-competitive small molecule inhibitor of casein kinase 2 (CK2) catalytic activity, suppresses replication of different HPV types, including novel HPV5NLuc, HPV11NLuc and HPV18NLuc marker genomes, but enhances the replication of HPV16 and HPV31. We further corroborate our findings using short interfering RNA (siRNA)-mediated knockdown of CK2 α and α' subunits in U2OS and CIN612 cells; we show that while both subunits are expressed in these cell lines, CK2α is required for HPV replication, but CK2α' is not. Furthermore, we demonstrate that CK2α acts in a kinase activity-dependent manner and regulates the stability and nuclear retention of endogenous E1 proteins of HPV11 and HPV18. This unique feature of CK2α makes it an attractive target for developing antiviral agents.


Subject(s)
Papillomaviridae/physiology , Papillomavirus Infections/virology , Phosphoproteins/metabolism , Viral Proteins/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/virology , Casein Kinase II/genetics , Casein Kinase II/metabolism , Humans , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/virology , Papillomavirus Infections/genetics , Papillomavirus Infections/metabolism , Phosphoproteins/genetics , Phosphorylation , Tumor Cells, Cultured , Viral Proteins/genetics
6.
Virology ; 514: 142-155, 2018 Jan 15.
Article in English | MEDLINE | ID: mdl-29179037

ABSTRACT

Nuclear myosin 1c (NM1) associates with RNA polymerases and is a partner in the chromatin remodeling complex B-WICH. This complex, which also contains WSTF and SNF2h proteins, is involved in transcriptional regulation. We report herein that papillomavirus protein E2 binds to NM1 and co-precipitates with the WSTF and SNF2h proteins. Our data suggest that E2 associates with the cellular B-WICH complex through binding to NM1. E2 and NM1 associate via their N-terminal domains and this interaction is ATP dependent. The cellular multifunctional protein Brd4 and beta-actin are also present in the NM1-E2 complex. NM1 downregulation by siRNA increases the replication of the BPV1 and HPV5 genomes but does not affect HPV18 genome replication. These results suggest that the B-WICH complex may play a role in the papillomavirus life cycle through NM1 and E2 protein interaction.


Subject(s)
Betapapillomavirus/metabolism , DNA-Binding Proteins/metabolism , Human papillomavirus 18/metabolism , Myosin Type I/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/metabolism , Virus Replication , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Betapapillomavirus/genetics , Bovine papillomavirus 1/genetics , Bovine papillomavirus 1/metabolism , Cell Cycle Proteins , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/genetics , Host-Pathogen Interactions , Human papillomavirus 18/chemistry , Human papillomavirus 18/genetics , Humans , Myosin Type I/chemistry , Myosin Type I/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Protein Binding , Protein Domains , Transcription Factors/genetics , Transcription Factors/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
7.
Nucleus ; 6(4): 289-300, 2015.
Article in English | MEDLINE | ID: mdl-26218798

ABSTRACT

Technological advantages in sequencing and proteomics have revealed the remarkable diversity of alternative protein isoforms. Typically, the localization and functions of these isoforms are unknown and cannot be predicted. Also the localization signals leading to particular subnuclear compartments have not been identified and thus, predicting alternative functions due to alternative subnuclear localization is limited only to very few subnuclear compartments. Knowledge of the localization and function of alternative protein isoforms allows for a greater understanding of cellular complexity. In this article, we characterize a short and well-defined signal targeting the bovine papillomavirus type 1 E8/E2 protein to the nuclear matrix. The targeting signal comprises the peptide coded by E8 ORF, which is spliced together with part of the E2 ORF to generate the E8/E2 mRNA. Localization to the nuclear matrix correlates well with the transcription repression activities of E8/E2; a single point mutation directs the E8/E2 protein into the nucleoplasm, and transcription repression activity is lost. Our data prove that adding as few as ˜10 amino acids by alternative transcription/alternative splicing drastically alters the function and subnuclear localization of proteins. To our knowledge, E8 is the shortest known nuclear matrix targeting signal.


Subject(s)
Bovine papillomavirus 1/genetics , DNA-Binding Proteins/genetics , Genome, Viral , Nuclear Matrix/genetics , Oncogene Proteins, Viral/genetics , Viral Proteins/genetics , Animals , CHO Cells , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Cricetulus , DNA-Binding Proteins/metabolism , Epigenetic Repression , Nuclear Matrix/metabolism , Oncogene Proteins, Viral/metabolism , Promoter Regions, Genetic , Transcription, Genetic , Viral Proteins/metabolism
8.
J Virol ; 88(2): 961-73, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24198410

ABSTRACT

Betapapillomavirus replication and transcription have not been studied in detail because of a lack of suitable cellular systems supporting human papillomavirus (HPV) genome replication. We have recently shown that the human osteosarcoma cell line U2OS provides a useful environment for the genome replication of many different HPVs, including the betapapillomaviruses HPV5 and HPV8. Using mutational analysis and complementation assay, we demonstrated herein that the viral early proteins E1 and E2 are viral transfactors that are necessary and sufficient for HPV5 genome replication. We also identified four HPV5 early promoter regions with transcription start sites (TSSs) at nucleotides (nt) 184/191, 460, 840, and 1254, respectively, and the HPV late promoter with a TSS at nt 7640. In addition, we mapped the HPV5 early polyadenylation cleavage sites via 3' rapid amplification of cDNA ends (3'RACE) to nt 4457 and 4475. In total, 14 different viral mRNA species, originating from the HPV5 genome, were mapped in U2OS cells during transient and stable replication. The main splicing donor and acceptor sites identified herein are consistent with the data previously obtained in HPV5-positive skin lesions. In addition, we identified novel E8 open reading frame (ORF)-containing transcripts (E8^E1C and E8^E2C) expressed from the HPV5 genome. Similar to several other papillomaviruses, the product of the E8^E2C mRNA acts as a repressor of viral genome replication.


Subject(s)
Betapapillomavirus/genetics , Genome, Viral , Transcription, Genetic , Virus Replication , Betapapillomavirus/physiology , Cell Line, Tumor , Chromosome Mapping , Gene Expression Regulation, Viral , Humans , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/virology , Promoter Regions, Genetic , Transcription Initiation Site
SELECTION OF CITATIONS
SEARCH DETAIL
...