11,275 results on '"VIRAL ENTRY"'
Search Results
102. Differences in endosomal Rab gene expression between positive and negative COVID-19 patients.
- Author
-
Atik, Nur, Wirawan, Farruqi, Amalia, Riezki, Khairani, Astrid Feinisa, and Pradini, Gita Widya
- Subjects
SARS-CoV-2 ,MANN Whitney U Test ,GENE expression ,ENDOCYTOSIS ,COVID-19 - Abstract
Objective: SARS CoV-2, the etiologic agent of coronavirus disease-2019 (COVID-19) is well-known to use ACE2 to begin internalization. Some viruses enter the host cell through the endocytosis process and involve some endocytosis proteins, such as the Rab family. However, the relationship between SARS CoV-2 infection with endocytic mRNA RAB5, RAB7, and RAB11B is unknown. This study aims to compare the expression of RAB5, RAB7, and RAB11B between positive and negative COVID-19 patient groups. Results: Both viral and human epithelial RNA Isolation and RT-PCR were performed from 249 samples. The genes expression was analysed using appropriate statistical tests. We found the Median (inter-quartile range/IQR) of RAB5, RAB7, and RAB11B expression among the COVID-19 patient group was 2.99 (1.88), 0.17 (0.47), 0.47 (1.49), and 1.60 (2.88), 1.05 (2.49), 1.10 (3.96) among control group respectively. We proceeded with Mann Whitney U Test and found that RAB5 expression was significantly increased (P < 0.001), and RAB7 and RAB11B expression was significantly decreased (P < 0.001 and P = 0.036) in the COVID-19 patient group compared to the control group. This first report showed significant differences in RAB5, RAB7, and RAB11B exist between COVID-19 positive and negative patients. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
103. SARS-CoV-2 Spike Furin Cleavage Site and S29 Basic Residues Modulate the Entry Process in a Host Cell-Dependent Manner.
- Author
-
Lavie, Muriel, Dubuisson, Jean, and Belouzard, Sandrine
- Subjects
- *
SARS-CoV-2 , *VIRAL envelopes , *VIRAL envelope proteins , *ANGIOTENSIN converting enzyme , *PROTEOLYTIC enzymes , *LIFE cycles (Biology) - Abstract
SARS-CoV-2 spike (S) envelope glycoprotein constitutes the main determinant of virus entry and the target of host immune response, thus being of great interest for antiviral research. It is constituted of S1 and S2 subunits, which are involved in ACE2 receptor binding and fusion between the viral envelope and host cell membrane, respectively. Induction of the fusion process requires S cleavage at the S1-S2 junction and the S29 site located upstream of the fusion peptide. Interestingly, the SARS-CoV-2 spike harbors a 4-residue insertion at the S1-S2 junction that is absent in its closest relatives and constitutes a polybasic motif recognized by furin-like proteases. In addition, the S29 site is characterized by the presence of conserved basic residues. Here, we sought to determine the importance of the furin cleavage site (FCS) and the S29 basic residues for S-mediated entry functions. We determined the impact of mutations introduced at these sites on S processing, fusogenic activity, and its ability to mediate entry in different cellular backgrounds. Strikingly, mutation phenotypes were highly dependent on the host cell background. We confirmed that although the FCS was not absolutely required for virus entry, it contributed to extending the fusogenic potential of S. Cleavage site mutations, as well as inhibition of furin protease activity, affected the cell surface expression of S in a host cell-dependent manner. Finally, inhibition of furin activity differentially affected SARS-CoV-2 virus infectivity in the tested host cells, thereby confirming the host cell-dependent effect of spike processing for the viral life cycle. IMPORTANCE SARS-CoV-2 is responsible for the current global pandemic that has resulted in several million deaths. As the key determinant of virus entry into host cells and the main target of host immune response, the spike glycoprotein constitutes an attractive target for therapeutics development. Entry functions of spike rely on its processing at two sites by host cell proteases. While SARS-CoV-2 spike differs from its closest relatives by the insertion of a basic furin cleavage motif at the first site, it harbors conserved basic residues at the second cleavage site. Characterization of the importance of the basic sequences present at the two cleavage sites revealed that they were influencing spike processing, intracellular localization, induction of fusion, and entry in a host cell-dependent manner. Thus, our results revealed a high heterogeneity in spike sequence requirement for entry functions in the different host cells, in agreement with the high adaptability of the SARS-CoV-2 virus. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
104. Development of a novel human CD147 knock-in NSG mouse model to test SARS-CoV-2 viral infection.
- Author
-
Badeti, Saiaditya, Jiang, Qingkui, Naghizadeh, Alireza, Tseng, Hsiang-chi, Bushkin, Yuri, Marras, Salvatore A. E., Nisa, Annuurun, Tyagi, Sanjay, Chen, Fei, Romanienko, Peter, Yehia, Ghassan, Evans, Deborah, Lopez-Gonzalez, Moises, Alland, David, Russo, Riccardo, Gause, William, Shi, Lanbo, and Liu, Dongfang
- Subjects
- *
LABORATORY mice , *ANIMAL disease models , *SARS-CoV-2 , *ANGIOTENSIN converting enzyme , *COVID-19 , *IMMUNOSTAINING , *VIRUS diseases , *FLOW cytometry - Abstract
Background: An animal model that can mimic the SARS-CoV-2 infection in humans is critical to understanding the rapidly evolving SARS-CoV-2 virus and for development of prophylactic and therapeutic strategies to combat emerging mutants. Studies show that the spike proteins of SARS-CoV and SARS-CoV-2 bind to human angiotensin-converting enzyme 2 (hACE2, a well-recognized, functional receptor for SARS-CoV and SARS-CoV-2) to mediate viral entry. Several hACE2 transgenic (hACE2Tg) mouse models are being widely used, which are clearly invaluable. However, the hACE2Tg mouse model cannot fully explain: (1) low expression of ACE2 observed in human lung and heart, but lung or heart failure occurs frequently in severe COVID-19 patients; (2) low expression of ACE2 on immune cells, but lymphocytopenia occurs frequently in COVID-19 patients; and (3) hACE2Tg mice do not mimic the natural course of SARS-CoV-2 infection in humans. Moreover, one of most outstanding features of coronavirus infection is the diversity of receptor usage, which includes the newly proposed human CD147 (hCD147) as a possible co-receptor for SARS-CoV-2 entry. It is still debatable whether CD147 can serve as a functional receptor for SARS-CoV-2 infection or entry. Results: Here we successfully generated a hCD147 knock-in mouse model (hCD147KI) in the NOD-scid IL2Rgammanull (NSG) background. In this hCD147KI-NSG mouse model, the hCD147 genetic sequence was placed downstream of the endogenous mouse promoter for mouse CD147 (mCD147), which creates an in vivo model that may better recapitulate physiological expression of hCD147 proteins at the molecular level compared to the existing and well-studied K18-hACE2-B6 (JAX) model. In addition, the hCD147KI-NSG mouse model allows further study of SARS-CoV-2 in the immunodeficiency condition which may assist our understanding of this virus in the context of high-risk populations in immunosuppressed states. Our data show (1) the human CD147 protein is expressed in various organs (including bronchiolar epithelial cells) in hCD147KI-NSG mice by immunohistochemical staining and flow cytometry; (2) hCD147KI-NSG mice are marginally sensitive to SARS-CoV-2 infection compared to WT-NSG littermates characterized by increased viral copies by qRT-PCR and moderate body weight decline compared to baseline; (3) a significant increase in leukocytes in the lungs of hCD147KI-NSG mice, compared to infected WT-NSG mice. Conclusions: hCD147KI-NSG mice are more sensitive to COVID-19 infection compared to WT-NSG mice. The hCD147KI-NSG mouse model can serve as an additional animal model for further interrogation whether CD147 serve as an independent functional receptor or accessory receptor for SARS-CoV-2 entry and immune responses. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
105. The Process of Filopodia Induction during HPV Infection.
- Author
-
Biondo, Alyssa and Meneses, Patricio I.
- Subjects
- *
FILOPODIA , *PAPILLOMAVIRUS diseases , *EPITHELIAL cells , *CELL membranes , *PAPILLOMAVIRUSES , *VIRUS diseases - Abstract
Human Papillomavirus 16 (HPV16) infects mucosal and epithelial cells and has been identified as a high-risk HPV type that is an etiologic agent of human cancers. The initial infectious process, i.e., the binding of the virus particle and its entry into the host cell, has been studied extensively, although it is not fully understood. There is still a gap in understanding the steps by which the virus is able to cross the plasma membrane after receptor binding. In this study, we demonstrate that after HPV16 comes into contact with a plasma membrane receptor, there are cytoskeletal changes resulting in an increase of filopodia numbers. This increase in filopodia numbers was transient and was maintained during the first two hours after virus addition. Our data show that there is a statistically significant increase in infection when filopodia numbers are increased by the addition of drug and virus simultaneously, and a decrease in virus infection when filopodia formation is inhibited. We describe that HPV16 binding results in the activation of Cdc42 GTPase that in turn results in an increase in filopodia. siRNA directed at Cdc42 GTPase resulted in a statistically significant reduction of infection and a corresponding lack of filopodia induction. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
106. Role of Serine Proteases and Host Cell Receptors Involved in Proteolytic Activation, Entry of SARS-CoV-2 and Its Current Therapeutic Options
- Author
-
Malik T and Dessie G
- Subjects
sars-cov-2 ,proteolytic activation ,viral entry ,Infectious and parasitic diseases ,RC109-216 - Abstract
Gashaw Dessie, Tabarak Malik Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, EthiopiaCorrespondence: Gashaw Dessie Tel +251 975152796Email dessiegashaw@yahoo.comAbstract: The current global pandemic of a novel severe acute respiratory syndrome coronavirus-2 continues with its public health disaster beginning from late December 2019 in Wuhan, Hubei province, China. The scientific community has tried to fight against this novel coronavirus through vaccine development and designing different candidate drugs. However, there is no well-defined therapy to prevent 2019-nCov infection, thus complete prevention of the virus remains difficult. Therefore, it is a critical factor for death of millions worldwide. Many clinical trials and insights are ongoing in the struggle with this pandemic of SARS-CoV-2. SARS-CoV-2 entry into the host cell requires host cell angiotensin-converting enzyme-2 (ACE2) and glucose regulated protein 78 (GRP78). On the other hand, proteolytic activation of the viral spike protein (S protein) needs the host cell serine proteases, including transmembrane serine protease 2 (TMPRSS2), cathepsins, trypsin and furin. This review focuses on the protein involved in the mechanism of entry, and proteolytic activation. In addition, it looks at current therapeutic options for SARS-CoV-2.Keywords: SARS-CoV-2, proteolytic activation, viral entry
- Published
- 2021
107. Insights into biological therapeutic strategies for COVID-19
- Author
-
Xiaolong Tian, Cheng Li, Yanling Wu, and Tianlei Ying
- Subjects
COVID-19 ,SARS-CoV-2 ,Biotherapeutics ,Viral entry ,Viral replication ,Cytokine storm ,Science (General) ,Q1-390 - Abstract
The worldwide pandemic of novel coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that emerged in late December 2019 requires the urgent development of therapeutic options. So far, numerous studies have investigated and uncovered the underlying epidemiology and clinical characteristics of COVID-19 infections in order to develop effective drugs. Compared with antiviral small-molecule inhibitors, biotherapeutics have unique advantages such as fewer side effects by virtue of their high specificity, and thus can be rapidly developed for promising treatments of COVID-19. Here, we summarize potential biotherapeutics and their mechanisms of action, including convalescent plasma, therapeutic antibodies, peptides, engineered ACE2, interferons, cytokine inhibitors, and RNAi-based therapeutics, and discuss in depth the advancements and precautions for each type of biotherapeutics in the treatment of COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
108. Genome-Wide Knockout Screen Identifies Human Sialomucin CD164 as an Essential Entry Factor for Lymphocytic Choriomeningitis Virus
- Author
-
Jamin Liu, Kristeene A. Knopp, Elze Rackaityte, Chung Yu Wang, Matthew T. Laurie, Sara Sunshine, Andreas S. Puschnik, and Joseph L. DeRisi
- Subjects
CD164 ,CRISPR screen ,lymphocytic choriomeningitis virus ,viral entry ,Microbiology ,QR1-502 - Abstract
ABSTRACT Lymphocytic choriomeningitis virus (LCMV) is a well-studied mammarenavirus that can be fatal in congenital infections. However, our understanding of LCMV and its interactions with human host factors remains incomplete. Here, host determinants affecting LCMV infection were investigated through a genome-wide CRISPR knockout screen in A549 cells, a human lung adenocarcinoma line. We identified and validated a variety of novel host factors that play a functional role in LCMV infection. Among these, knockout of the sialomucin CD164, a heavily glycosylated transmembrane protein, was found to ablate infection with multiple LCMV strains but not other hemorrhagic mammarenaviruses in several cell types. Further characterization revealed a dependency of LCMV entry on the cysteine-rich domain of CD164, including an N-linked glycosylation site at residue 104 in that region. Given the documented role of LCMV with respect to transplacental human infections, CD164 expression was investigated in human placental tissue and placental cell lines. CD164 was found to be highly expressed in the cytotrophoblast cells, an initial contact site for pathogens within the placenta, and LCMV infection in placental cells was effectively blocked using a monoclonal antibody specific to the cysteine-rich domain of CD164. Together, this study identifies novel factors associated with LCMV infection of human tissues and highlights the importance of CD164, a sialomucin that previously had not been associated with viral infection. IMPORTANCE Lymphocytic choriomeningitis virus (LCMV) is a human-pathogenic mammarenavirus that can be fatal in congenital infections. Although frequently used in the study of persistent infections in the field of immunology, aspects of this virus’s life cycle remain incomplete. For example, while viral entry has been shown to depend on a cell adhesion molecule, DAG1, genetic knockout of this gene allows for residual viral infection, implying that additional receptors can mediate cell entry. The significance of our study is the identification of host factors important for successful infection, including the sialomucin CD164, which had not been previously associated with viral infection. We demonstrated that CD164 is essential for LCMV entry into human cells and can serve as a possible therapeutic target for treatment of congenital infection.
- Published
- 2022
- Full Text
- View/download PDF
109. Receptor Activation of HIV-1 Env Leads to Asymmetric Exposure of the gp41 Trimer
- Author
-
Trkola, Alexandra [Thomas Jefferson Univ., Philadelphia, PA (United States). Sidney Kimmel Cancer Center, Dept. of Biochemistry and Molecular Biology]
- Published
- 2016
- Full Text
- View/download PDF
110. Structural basis for norovirus neutralization by an HBGA blocking human IgA antibody
- Author
-
Shanker, Sreejesh, Czakó, Rita, Sapparapu, Gopal, Alvarado, Gabriela, Viskovska, Maria, Sankaran, Banumathi, Atmar, Robert L, Crowe, James E, Estes, Mary K, and Prasad, BV Venkataram
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Foodborne Illness ,Immunization ,Prevention ,Digestive Diseases ,Biotechnology ,Vaccine Related ,Biodefense ,Amino Acid Sequence ,Antibodies ,Blocking ,Antigens ,Blood Group Antigens ,Crystallography ,X-Ray ,Epitopes ,Genotype ,Humans ,Immunoglobulin A ,Immunoglobulin Fab Fragments ,Models ,Molecular ,Neutralization Tests ,Norovirus ,Protein Domains ,Viral Proteins ,norovirus ,HBGA-blockade antibody ,crystal structure ,antibody neutralization ,viral entry - Abstract
Human noroviruses (HuNoVs) cause sporadic and epidemic gastroenteritis worldwide. They are classified into two major genogroups (GI and GII), with each genogroup further divided into multiple genotypes. Susceptibility to these viruses is influenced by genetically determined histo-blood group antigen (HBGA) expression. HBGAs function as cell attachment factors by binding to a surface-exposed region in the protruding (P) domain of the capsid protein. Sequence variations in this region that result in differential HBGA binding patterns and antigenicity are suggested to form a basis for strain diversification. Recent studies show that serum antibodies that block HBGA binding correlate with protection against illness. Although genogroup-dependent variation in HBGA binding specificity is structurally well characterized, an understanding of how antibodies block HBGA binding and how genotypic variations affect such blockade is lacking. Our crystallographic studies of the GI.1 P domain in complex with the Fab fragment of a human IgA monoclonal antibody (IgA 5I2) with HBGA blocking activity show that the antibody recognizes a conformational epitope formed by two surface-exposed loop clusters in the P domain. The antibody engulfs the HBGA binding site but does not affect its structural integrity. An unusual feature of the antigen recognition by IgA 5I2 is the predominant involvement of the CDR light chain 1 in contrast to the commonly observed CDR heavy chain 3, providing a unique perspective into antibody diversity in antigen recognition. Identification of the antigenic site in the P domain shows how genotypic variations might allow escape from antibody neutralization and exemplifies the interplay between antigenicity and HBGA specificity in HuNoV evolution.
- Published
- 2016
111. Structure-Based Development of SARS-CoV-2 Spike Interactors.
- Author
-
Squeglia, Flavia, Romano, Maria, Esposito, Luciana, Barra, Giovanni, Campiglia, Pietro, Sala, Marina, Scala, Maria Carmina, Ruggiero, Alessia, and Berisio, Rita
- Subjects
- *
SARS-CoV-2 Delta variant , *SARS-CoV-2 , *MOLECULAR dynamics , *ANGIOTENSIN converting enzyme , *COVID-19 pandemic - Abstract
Coronaviruses, including SARS-CoV-2 (the etiological agent of the current COVID-19 pandemic), rely on the surface spike glycoprotein to access the host cells, mainly through the interaction of their receptor-binding domain (RBD) with the human angiotensin-converting enzyme 2 (ACE2). Therefore, molecular entities able to interfere with the binding of the SARS-CoV-2 spike protein to ACE2 have great potential to inhibit viral entry. Starting from the available structural data on the interaction between SARS-CoV-2 spike protein and the host ACE2 receptor, we engineered a set of soluble and stable spike interactors, here denoted as S-plugs. Starting from the prototype S-plug, we adopted a computational approach by combining stability prediction, associated to single-point mutations, with molecular dynamics to enhance both S-plug thermostability and binding affinity to the spike protein. The best developed molecule, S-plug3, possesses a highly stable α-helical con-formation (with melting temperature Tm of 54 °C) and can interact with the spike RBD and S1 domains with similar low nanomolar affinities. Importantly, S-plug3 exposes the spike RBD to almost the same interface as the human ACE2 receptor, aimed at the recognition of all ACE2-accessing coronaviruses. Consistently, S-plug3 preserves a low nanomolar dissociation constant with the delta B.1.617.2 variant of SARS-CoV-2 spike protein (KD = 29.2 ± 0.6 nM). Taken together, we provide valid starting data for the development of therapeutical and diagnostic tools against coronaviruses accessing through ACE2. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
112. Glycopeptide Antibiotic Teicoplanin Inhibits Cell Entry of SARS-CoV-2 by Suppressing the Proteolytic Activity of Cathepsin L.
- Author
-
Yu, Fei, Pan, Ting, Huang, Feng, Ying, Ruosu, Liu, Jun, Fan, Huimin, Zhang, Junsong, Liu, Weiwei, Lin, Yingtong, Yuan, Yaochang, Yang, Tao, Li, Rong, Zhang, Xu, Lv, Xi, Chen, Qianyu, Liang, Anqi, Zou, Fan, Liu, Bingfeng, Hu, Fengyu, and Tang, Xiaoping
- Subjects
TEICOPLANIN ,SARS-CoV-2 ,GLYCOPEPTIDE antibiotics ,ANTIBIOTICS ,COVID-19 ,EBOLA virus - Abstract
Since the outbreak of the coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), public health worldwide has been greatly threatened. The development of an effective treatment for this infection is crucial and urgent but is hampered by the incomplete understanding of the viral infection mechanisms and the lack of specific antiviral agents. We previously reported that teicoplanin, a glycopeptide antibiotic that has been commonly used in the clinic to treat bacterial infection, significantly restrained the cell entry of Ebola virus, SARS-CoV, and MERS-CoV by specifically inhibiting the activity of cathepsin L (CTSL). Here, we found that the cleavage sites of CTSL on the spike proteins of SARS-CoV-2 were highly conserved among all the variants. The treatment with teicoplanin suppressed the proteolytic activity of CTSL on spike and prevented the cellular infection of different pseudotyped SARS-CoV-2 viruses. Teicoplanin potently prevented the entry of SARS-CoV-2 into the cellular cytoplasm with an IC
50 of 2.038 μM for the Wuhan-Hu-1 reference strain and an IC50 of 2.116 μM for the SARS-CoV-2 (D614G) variant. The pre-treatment of teicoplanin also prevented SARS-CoV-2 infection in hACE2 mice. In summary, our data reveal that CTSL is required for both SARS-CoV-2 and SARS-CoV infection and demonstrate the therapeutic potential of teicoplanin for universal anti-CoVs intervention. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
113. Salvia miltiorrhiza Bunge as a Potential Natural Compound against COVID-19.
- Author
-
Petitjean, Simon J. L., Lecocq, Marylène, Lelong, Camille, Denis, Robin, Defrère, Sylvie, Mariage, Pierre-Antoine, Alsteens, David, and Pilette, Charles
- Subjects
- *
SALVIA miltiorrhiza , *ATOMIC force microscopy , *COVID-19 , *CHINESE medicine , *COVID-19 treatment - Abstract
Salvia miltiorrhiza Bunge, commonly called danshen, is widely used in traditional Chinese medicine for its cardiovascular and neuroprotective effects, which include antioxidative, anti-inflammatory, and antifibrotic properties. The purpose of this study was to evaluate the preclinical potential of S. miltiorrhiza extracts for the treatment of COVID-19. First, the impact of the extract on the binding between SARS-CoV-2 and the cellular ACE2 receptors was assessed using atomic force microscopy (AFM), showing a significant reduction in binding by the extract at concentrations in the µg/mL range. Second, the interference of this extract with the inflammatory response of blood mononuclear cells (PBMCs) was determined, demonstrating potent inhibitory properties in the same concentration range on pro-inflammatory cytokine release and interference with the activation of NFκB signaling. Together, these in vitro data demonstrate the potential of S. miltiorrhiza against COVID-19, consisting first of the blockade of the binding of SARS-CoV-2 to the ACE2 receptor and the mitigation of the inflammatory response from leukocytes by interfering with NFκB signaling. This dataset prompts the launch of a clinical trial to address in vivo the clinical benefits of this promising agent. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
114. Distinctive Roles of Furin and TMPRSS2 in SARS-CoV-2 Infectivity.
- Author
-
Essalmani, Rachid, Jain, Jaspreet, Susan-Resiga, Delia, Andréo, Ursula, Evagelidis, Alexandra, Derbali, Rabeb Mouna, Huynh, David N., Dallaire, Frédéric, Laporte, Mélanie, Delpal, Adrien, Sutto-Ortiz, Priscila, Coutard, Bruno, Mapa, Claudine, Wilcoxen, Keith, Decroly, Etienne, Pham, Tram N.Q., Cohen, Éric A., and Seidah, Nabil G.
- Subjects
- *
SARS-CoV-2 , *CHIMERIC proteins - Abstract
The spike protein (S) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directs infection of the lungs and other tissues following its binding to the angiotensin-converting enzyme 2 (ACE2) receptor. For effective infection, the S protein is cleaved at two sites: S1/S2 and S29. The “priming” of the surface S protein at S1/S2 (PR...RAR...685↓) [the underlined basic amino acids refer to critical residues needed for the furin recognition] by furin has been shown to be important for SARS-CoV-2 infectivity in cells and small-animal models. In this study, for the first time we unambiguously identified by proteomics the fusion activation site S29 as KPSKR...815↓ (the underlined basic amino acids refer to critical residues needed for the furin recognition) and demonstrated that this cleavage was strongly enhanced by ACE2 engagement with the S protein. Novel pharmacological furin inhibitors (BOS inhibitors) effectively blocked endogenous S protein processing at both sites in HeLa cells, and SARS-CoV-2 infection of lung-derived Calu-3 cells was completely prevented by combined inhibitors of furin (BOS) and type II transmembrane serine protease 2 (TMPRSS2) (camostat). Quantitative analyses of cell-tocell fusion and S protein processing revealed that ACE2 shedding by TMPRSS2 was required for TMPRSS2-mediated enhancement of fusion in the absence of S1/S2 priming. We further demonstrated that the collectrin dimerization domain of ACE2 was essential for the effect of TMPRSS2 on cell-to-cell fusion. Overall, our results indicate that furin and TMPRSS2 act synergistically in viral entry and infectivity, supporting the combination of furin and TMPRSS2 inhibitors as potent antivirals against SARS-CoV-2. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
115. Identification of Entry Inhibitors against Delta and Omicron Variants of SARS-CoV-2.
- Author
-
Lee, Richard Kuan-Lin, Li, Tian-Neng, Chang, Sui-Yuan, Chao, Tai-Ling, Kuo, Chun-Hsien, Pan, Max Yu-Chen, Chiou, Yu-Ting, Liao, Kuan-Ju, Yang, Yi, Wu, Yi-Hsuan, Huang, Chen-Hao, Juan, Hsueh-Fen, Hsieh, Hsing-Pang, and Wang, Lily Hui-Ching
- Subjects
- *
COVID-19 , *SARS-CoV-2 Omicron variant , *SARS-CoV-2 Delta variant , *SARS-CoV-2 - Abstract
Entry inhibitors against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are urgently needed to control the outbreak of coronavirus disease 2019 (COVID-19). This study developed a robust and straightforward assay that detected the molecular interaction between the receptor-binding domain (RBD) of viral spike protein and the angiotensin-converting enzyme 2 (ACE2) receptor in just 10 min. A drug library of 1068 approved compounds was used to screen for SARS-CoV2 entry inhibition, and 9 active drugs were identified as specific pseudovirus entry inhibitors. A plaque reduction neutralization test using authentic SARS-CoV-2 virus in Vero E6 cells confirmed that 2 of these drugs (Etravirine and Dolutegravir) significantly inhibited the infection of SARS-CoV-2. With molecular docking, we showed that both Etravirine and Dolutegravir are preferentially bound to primary ACE2-interacting residues on the RBD domain, implying that these two drug blocks may prohibit the viral attachment of SARS-CoV-2. We compared the neutralizing activities of these entry inhibitors against different pseudoviruses carrying spike proteins from alpha, beta, gamma, and delta variants. Both Etravirine and Dolutegravir showed similar neutralizing activities against different variants, with EC50 values between 4.5 to 5.8 nM for Etravirine and 10.2 to 22.9 nM for Dolutegravir. These data implied that Etravirine and Dolutegravir may serve as general spike inhibitors against dominant viral variants of SARS-CoV-2. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
116. Human Papillomavirus L2 Capsid Protein Stabilizes γ-Secretase during Viral Infection.
- Author
-
Crite, Mac and DiMaio, Daniel
- Subjects
- *
VIRUS diseases , *PRESENILINS , *PAPILLOMAVIRUSES , *PROTEINS - Abstract
Intracellular trafficking of human papillomavirus (HPV) during virus entry requires γ-secretase, a cellular protease consisting of a complex of four cellular transmembrane (TM) proteins. γ-secretase typically cleaves substrate proteins but it plays a non-canonical role during HPV entry. γ-secretase binds to the HPV minor capsid protein L2 and facilitates its insertion into the endosomal membrane. After insertion, L2 protrudes into the cytoplasm, which allows HPV to bind other cellular factors required for proper virus trafficking into the retrograde transport pathway. Here, we further characterize the interaction between γ-secretase and HPV L2. We show that γ-secretase is required for cytoplasmic protrusion of L2 and that L2 associates strongly with the PS1 catalytic subunit of γ-secretase and stabilizes the γ-secretase complex. Mutational studies revealed that a putative TM domain in HPV16 L2 cannot be replaced by a foreign TM domain, that infectivity of HPV TM mutants is tightly correlated with γ-secretase binding and stabilization, and that the L2 TM domain is required for protrusion of the L2 protein into the cytoplasm. These results provide new insight into the interaction between γ-secretase and L2 and highlight the importance of the native HPV L2 TM domain for proper virus trafficking during entry. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
117. Pseudorabies Virus Inhibits Expression of Liver X Receptors to Assist Viral Infection.
- Author
-
Wang, Yi, Li, Guo-Li, Qi, Yan-Li, Li, Li-Yun, Wang, Lu-Fang, Wang, Cong-Rong, Niu, Xin-Rui, Liu, Tao-Xue, Wang, Jiang, Yang, Guo-Yu, Zeng, Lei, and Chu, Bei-Bei
- Subjects
- *
AUJESZKY'S disease virus , *VIRUS diseases , *LIVER , *CLASSICAL swine fever , *CHOLESTEROL metabolism , *CHOLESTEROL - Abstract
Pseudorabies virus (PRV) is a contagious herpesvirus that causes Aujeszky's disease and economic losses worldwide. Liver X receptors (LXRs) belong to the nuclear receptor superfamily and are critical for the control of lipid homeostasis. However, the role of LXR in PRV infection has not been fully established. In this study, we found that PRV infection downregulated the mRNA and protein levels of LXRα and LXRβ in vitro and in vivo. Furthermore, we discovered that LXR activation suppressed PRV proliferation, while LXR inhibition promoted PRV proliferation. We demonstrated that LXR activation-mediated reduction of cellular cholesterol was critical for the dynamics of PRV entry-dependent clathrin-coated pits. Replenishment of cholesterol restored the dynamics of clathrin-coated pits and PRV entry under LXR activation conditions. Interestingly, T0901317, an LXR agonist, prevented PRV infection in mice. Our results support a model that PRV modulates LXR-regulated cholesterol metabolism to facilitate viral proliferation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
118. Role of Protein–Lipid Interactions in Viral Entry.
- Author
-
Nieto‐Garai, Jon Ander, Contreras, Francesc‐Xabier, Arboleya, Aroa, and Lorizate, Maier
- Subjects
PROTEIN-lipid interactions ,MEMBRANE lipids ,LIPID rafts ,LIPIDS - Abstract
The viral entry consists of several sequential events that ensure the attachment of the virus to the host cell and the introduction of its genetic material for the continuation of the replication cycle. Both cellular and viral lipids have gained a wider focus in recent years in the field of viral entry, as they are found to play key roles in different steps of the process. The specific role is summarized that lipids and lipid membrane nanostructures play in viral attachment, fusion, and immune evasion and how they can be targeted with antiviral therapies. Finally, some of the limitations of techniques commonly used for protein–lipid interactions studies are discussed, and new emerging tools are reviewed that can be applied to this field. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
119. Coronaviruses fusion with the membrane and entry to the host cell
- Author
-
Ewelina Wędrowska, Tomasz Wandtke, Tomasz Senderek, Elżbieta Piskorska, and Piotr Kopiński
- Subjects
coronavirus ,spike protein ,membrane fusion ,viral entry ,nonstructural proteins ,replication complex ,Agriculture ,Environmental sciences ,GE1-350 - Abstract
Coronaviruses (CoVs) are positive-strand RNA viruses with the largest genome among all RNA viruses. They are able to infect many host, such as mammals or birds. Whereas CoVs were identified 1930s, they became known again in 2003 as the agents of the Severe Acute Respiratory Syndrome (SARS). The spike protein is thought to be essential in the process of CoVs entry, because it is associated with the binding to the receptor on the host cell. It is also involved in cell tropism and pathogenesis. Receptor recognition is the crucial step in the infection. CoVs are able to bind a variety of receptors, although the selection of receptor remains unclear. Coronaviruses were initially believed to enter cells by fusion with the plasma membrane. Further studies demonstrated that many of them involve endocytosis through clathrin-dependent, caveolae-dependent, clathrin-independent, as well as caveolae-independent mechanisms. The aim of this review is to summarise current knowledge about coronaviruses, focussing especially on CoVs entry into the host cell. Advances in understanding coronaviruses replication strategy and the functioning of the replicative structures are also highlighted. The development of host-directed antiviral therapy seems to be a promising way to treat infections with SARS-CoV or other pathogenic coronaviruses. There is still much to be discovered in the inventory of pro- and anti-viral host factors relevant for CoVs replication. The latest pandemic danger, originating from China, has given our previously prepared work even more of topicality.
- Published
- 2020
- Full Text
- View/download PDF
120. Identification of the dietary supplement capsaicin as an inhibitor of Lassa virus entry
- Author
-
Ke Tang, Xiaoyu Zhang, and Ying Guo
- Subjects
Lassa virus ,Fusion inhibitor ,Capsaicin ,Dietary supplement ,Natural product antivirals ,Viral entry ,Therapeutics. Pharmacology ,RM1-950 - Abstract
The limited treatment options for the increasing occurrence of Lassa hemorrhagic fever in West Africa poses an urgent need for the discovery and development of novel therapeutics. Dietary supplements, especially natural products that are edible and safe for human use, are a good source of drug discovery with potential for uncovering novel applications. In this study, we tested 40 natural products of dietary supplements and identified capsaicin, a common dietary supplement abundant in chili peppers, as an inhibitor of Lassa virus (LASV) entry with EC50 of 6.9–10.0 μmol/L using an HIV based pseudovirus platform. Capsaicin inhibits the entry of five LASV strains but not against the Old World arenavirus lymphocytic choriomeningitis virus (LCMV), showing a preferential activity against LASV. Capsaicin inhibits LASV entry by blocking the pH dependent viral fusion through affecting the stable signal peptide (SSP)-GP2 transmembrane (GP2TM) region of the LASV surface glycoprotein. Mutational study revealed the key residues Ala25, Val431, Phe434 and Val435 in SSP-GP2TM region in capsaicin's antiviral effect. This study for the first time reveals a direct acting antiviral effect of capsaicin against the hemorrhagic fever causing LASV, providing detailed interaction hot spots in the unique SSP-GP2TM interface of LASV glycoprotein that is crucial in fusion inhibition, and offering a new strategy in discovering and developing antivirals from natural products that are safe for human use.
- Published
- 2020
- Full Text
- View/download PDF
121. Glycopeptide Antibiotic Teicoplanin Inhibits Cell Entry of SARS-CoV-2 by Suppressing the Proteolytic Activity of Cathepsin L
- Author
-
Fei Yu, Ting Pan, Feng Huang, Ruosu Ying, Jun Liu, Huimin Fan, Junsong Zhang, Weiwei Liu, Yingtong Lin, Yaochang Yuan, Tao Yang, Rong Li, Xu Zhang, Xi Lv, Qianyu Chen, Anqi Liang, Fan Zou, Bingfeng Liu, Fengyu Hu, Xiaoping Tang, Linghua Li, Kai Deng, Xin He, Hui Zhang, Yiwen Zhang, and Xiancai Ma
- Subjects
teicoplanin ,SARS-CoV-2 ,spike ,cathepsin L ,viral entry ,Microbiology ,QR1-502 - Abstract
Since the outbreak of the coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), public health worldwide has been greatly threatened. The development of an effective treatment for this infection is crucial and urgent but is hampered by the incomplete understanding of the viral infection mechanisms and the lack of specific antiviral agents. We previously reported that teicoplanin, a glycopeptide antibiotic that has been commonly used in the clinic to treat bacterial infection, significantly restrained the cell entry of Ebola virus, SARS-CoV, and MERS-CoV by specifically inhibiting the activity of cathepsin L (CTSL). Here, we found that the cleavage sites of CTSL on the spike proteins of SARS-CoV-2 were highly conserved among all the variants. The treatment with teicoplanin suppressed the proteolytic activity of CTSL on spike and prevented the cellular infection of different pseudotyped SARS-CoV-2 viruses. Teicoplanin potently prevented the entry of SARS-CoV-2 into the cellular cytoplasm with an IC50 of 2.038 μM for the Wuhan-Hu-1 reference strain and an IC50 of 2.116 μM for the SARS-CoV-2 (D614G) variant. The pre-treatment of teicoplanin also prevented SARS-CoV-2 infection in hACE2 mice. In summary, our data reveal that CTSL is required for both SARS-CoV-2 and SARS-CoV infection and demonstrate the therapeutic potential of teicoplanin for universal anti-CoVs intervention.
- Published
- 2022
- Full Text
- View/download PDF
122. Senecavirus A Entry Into Host Cells Is Dependent on the Cholesterol-Mediated Endocytic Pathway
- Author
-
Meiyu Jia, Mingxia Sun, Yan-Dong Tang, Yu-Yuan Zhang, Haiwei Wang, Xuehui Cai, and Fandan Meng
- Subjects
SVA ,reporter virus ,viral entry ,pig ANTXR1 ,cholesterol ,Veterinary medicine ,SF600-1100 - Abstract
Senecavirus A (SVA), an important member of the Picornaviridae family, causes vesicular disease in pigs. Here, we generated an EGFP-expressing recombinant SVA re-SVA-EGFP, which exhibited similar growth kinetics to its parental virus. The reporter SVA was used to study the role of pig ANTXR1 (pANTXR1) in SVA infection in a porcine alveolar macrophage cell line (PAM-Tang cells). Knockdown of the pANTXR1 significantly reduced SVA infection and replication in PAM-Tang cells, while re-expression of the pANTXR1 promoted the cell susceptibility to SVA infection. The results indicated that pANTXR1 is a crucial receptor mediating SVA infection. Subsequently, the viral endocytosis pathways for SVA entry into pig cells were investigated and the results showed that cholesterol played an essential role in receptor-mediated SVA entry. Together, these results demonstrated that SVA entered into host cells through the pANTXR1-mediated cholesterol pathway. Our findings provide potential targets to develop antiviral drugs for the prevention of SVA infection in the pig population.
- Published
- 2022
- Full Text
- View/download PDF
123. Editorial: Significance of Cellular Lipids for Viral Replication and Pathogenesis
- Author
-
Ulrich Desselberger, Carolina Henritta Pohl, and Hester Gertruida O’Neill
- Subjects
lipid droplets ,sphingolipids ,cholesterol ,viral replication ,viral entry ,broadspectrum antiviral therapy ,Physiology ,QP1-981 - Published
- 2022
- Full Text
- View/download PDF
124. Lipoprotein receptors: A little grease for enveloped viruses to open the lock?
- Author
-
Cosset FL and Denolly S
- Subjects
- Humans, Animals, Flaviviridae metabolism, Virus Internalization, Receptors, Lipoprotein metabolism, Receptors, Lipoprotein genetics
- Abstract
Several studies recently highlighted the role of lipoprotein receptors in viral entry. These receptors are evolutionarily ancient proteins, key for the transport of lipids as well as other signaling molecules across the plasma membrane. Here, we discuss the different families of lipoprotein receptors and how they are hijacked by enveloped viruses to promote their entry into infected cells. While the usage of lipoprotein receptors was known for members of the Flaviviridae family and vesicular stomatitis virus, the last 4 years have seen the discovery that these receptors are used by many genetically unrelated viruses. We also emphasize how viral particles interact with these receptors and the possible targeting of these host factors as antiviral strategies., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
125. Viral entry mechanisms: the role of molecular simulation in unlocking a key step in viral infections.
- Author
-
Valério M, Buga CC, Melo MN, Soares CM, and Lousa D
- Abstract
Viral infections are a major global health concern, affecting millions of people each year. Viral entry is one of the crucial stages in the infection process, but its details remain elusive. Enveloped viruses are enclosed by a lipid membrane that protects their genetic material and these viruses are linked to various human illnesses, including influenza, and COVID-19. Due to the advancements made in the field of molecular simulation, significant progress has been made in unraveling the dynamic processes involved in viral entry of enveloped viruses. Simulation studies have provided deep insight into the function of the proteins responsible for attaching to the host receptors and promoting membrane fusion (fusion proteins), deciphering interactions between these proteins and receptors, and shedding light on the functional significance of key regions, such as the fusion peptide. These studies have already significantly contributed to our understanding of this critical aspect of viral infection and assisted the development of effective strategies to combat viral diseases and improve global health. This review focuses on the vital role of fusion proteins in facilitating the entry process of enveloped viruses and highlights the contributions of molecular simulation studies to uncover the molecular details underlying their mechanisms of action., (© 2024 The Author(s). FEBS Open Bio published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
126. Allosteric mechanism of membrane fusion activation in a herpesvirus.
- Author
-
González-Del Pino GL, Walsh RM Jr, Atanasiu D, Cairns TM, Saw WT, Cohen GH, and Heldwein EE
- Abstract
Herpesviridae infect nearly all humans for life, causing diseases that range from painful to life-threatening
1 . These viruses penetrate cells by employing a complex apparatus composed of separate receptor-binding, signal-transmitting, and membrane-fusing components2 . But how these components coordinate their functions is unknown. Here, we determined the 4.19-angstrom cryoEM reconstruction of the central signal-transmitting component from herpes simplex virus 2, the gH/gL complex, in its elusive pre-activation state. Analysis of the continuum of conformational ensembles observed in cryoEM data revealed a series of structural rearrangements in gH/gL that allosterically transmit the fusion-triggering signal from the receptor-binding glycoprotein gD to the membrane fusogen gB. Furthermore, we identified a structural "switch" element in gH/gL that refolds and flips 180 degrees during the transition from pre-activation to activated form. Conservation of this "switch" in gH/gL homologs suggests that the proposed fusion triggering mechanism may apply to all Herpesviridae and points to a new target for subunit-based vaccines and treatment efforts., Competing Interests: Competing interests: Authors declare that they have no competing interests.- Published
- 2024
- Full Text
- View/download PDF
127. Emerging New Therapies for Viral Hepatitis
- Author
-
Su, Pei-Yi, Shih, Chiaho, Chang, Mei-Hwei, editor, and Schwarz, Kathleen B., editor
- Published
- 2019
- Full Text
- View/download PDF
128. Conformation of HIV-1 Envelope Governs Rhesus CD4 Usage and Simian-Human Immunodeficiency Virus Replication
- Author
-
Geraldine Vilmen, Anna C. Smith, Hector Cervera Benet, Rajni Kant Shukla, Ross C. Larue, Alon Herschhorn, and Amit Sharma
- Subjects
HIV-1 envelope ,CD4 ,rhesus macaque ,SHIV ,conformation ,viral entry ,Microbiology ,QR1-502 - Abstract
ABSTRACT Infection of rhesus macaques with simian-human immunodeficiency viruses (SHIVs) is the preferred model system for vaccine development because SHIVs encode human immunodeficiency virus type 1 (HIV-1) envelope glycoproteins (Envs)—a key target of HIV-1 neutralizing antibodies. Since the goal of vaccines is to prevent new infections, SHIVs encoding circulating HIV-1 Env are desired as challenge viruses. Development of such biologically relevant SHIVs has been challenging, as they fail to infect rhesus macaques, mainly because most circulating HIV-1 Envs do not use rhesus CD4 (rhCD4) receptor for viral entry. Most primary HIV-1 Envs exist in a closed conformation and occasionally transit to a downstream, open conformation through an obligate intermediate conformation. Here, we provide genetic evidence that open Env conformations can overcome the rhCD4 entry barrier and increase replication of SHIVs in rhesus lymphocytes. Consistent with prior studies, we found that circulating HIV-1 Envs do not use rhCD4 efficiently for viral entry. However, by using HIV-1 Envs with single amino acid substitutions that alter their conformational state, we found that transitions to intermediate and open Env conformations allow usage of physiological levels of rhCD4 for viral entry. We engineered these single amino acid substitutions in the transmitted/founder HIV-1BG505 Envs encoded by SHIV-BG505 and found that open Env conformation enhances SHIV replication in rhesus lymphocytes. Lastly, CD4-mediated SHIV pulldown, sensitivity to soluble CD4, and fusogenicity assays indicated that open Env conformation promotes efficient rhCD4 binding and viral-host membrane fusion. These findings identify the conformational state of HIV-1 Env as a major determinant for rhCD4 usage, viral fusion, and SHIV replication. IMPORTANCE Rhesus macaques are a critical animal model for preclinical testing of HIV-1 vaccine and prevention approaches. However, HIV-1 does not replicate in rhesus macaques, and thus, chimeric simian-human immunodeficiency viruses (SHIVs), which encode HIV-1 envelope glycoproteins (Envs), are used as surrogate challenge viruses to infect rhesus macaques for modeling HIV-1 infection. Development of SHIVs encoding Envs from clinically relevant, circulating HIV-1 variants has been extremely challenging, as such SHIVs replicate poorly, if at all, in rhesus lymphocytes. This is most probably because many circulating HIV-1 Envs do not use rhesus CD4 efficiently for viral entry. In this study, we identified conformational state of HIV-1 envelope as a key determinant for rhesus CD4 usage, viral-host membrane fusion, and SHIV replication in rhesus lymphocytes.
- Published
- 2022
- Full Text
- View/download PDF
129. Targeting the Fusion Process of SARS-CoV-2 Infection by Small Molecule Inhibitors
- Author
-
Seung Bum Park, Parker Irvin, Zongyi Hu, Mohsin Khan, Xin Hu, Qiru Zeng, Catherine Chen, Miao Xu, Madeleine Leek, Ruochen Zang, James Brett Case, Wei Zheng, Siyuan Ding, and T. Jake Liang
- Subjects
SARS-CoV-2 ,COVID-19 ,viral entry ,viral fusion ,fusion inhibitor ,broad-spectrum antiviral ,Microbiology ,QR1-502 - Abstract
ABSTRACT Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a serious threat to global public health, underscoring the urgency of developing effective therapies. Therapeutics and, more specifically, direct-acting antiviral development are still very much in their infancy. Here, we report that two hepatitis C virus (HCV) fusion inhibitors identified in our previous study, dichlorcyclizine and fluoxazolevir, broadly block human coronavirus entry into various cell types. Both compounds were effective against various human-pathogenic CoVs in multiple assays based on vesicular stomatitis virus (VSV) pseudotyped with the spike protein and spike-mediated syncytium formation. The antiviral effects were confirmed in SARS-CoV-2 infection systems. These compounds were equally effective against recently emerged variants, including the delta variant. Cross-linking experiments and structural modeling suggest that the compounds bind to a hydrophobic pocket near the fusion peptide of S protein, consistent with their potential mechanism of action as fusion inhibitors. In summary, these fusion inhibitors have broad-spectrum antiviral activities and may be promising leads for treatment of SARS-CoV-2, its variants, and other pathogenic CoVs. IMPORTANCE SARS-CoV-2 is an enveloped virus that requires membrane fusion for entry into host cells. Since the fusion process is relatively conserved among enveloped viruses, we tested our HCV fusion inhibitors, dichlorcyclizine and fluoxazolevir, against SARS-CoV-2. We performed in vitro assays and demonstrated their effective antiviral activity against SARS-CoV-2 and its variants. Cross-linking experiments and structural modeling suggest that the compounds bind to a hydrophobic pocket in spike protein to exert their inhibitory effect on the fusion step. These data suggest that both dichlorcyclizine and fluoxazolevir are promising candidates for further development as treatment for SARS-CoV-2.
- Published
- 2022
- Full Text
- View/download PDF
130. Susceptibility of Human and Murine Dermal Fibroblasts to Herpes Simplex Virus 1 in the Absence and Presence of Extracellular Matrix.
- Author
-
Wirtz, Lisa, De La Cruz, Nydia C., Möckel, Maureen, and Knebel-Mörsdorf, Dagmar
- Subjects
- *
EXTRACELLULAR matrix , *FIBROBLASTS , *DERMIS - Abstract
Herpes simplex virus 1 (HSV-1) invades its human host via the skin and mucosa and initiates infection in the epithelium. While human and murine epidermis are highly susceptible to HSV-1, we recently observed rare infected cells in the human dermis and only minor infection efficiency in murine dermis upon ex vivo infection. Here, we investigated why cells in the dermis are so inefficiently infected and explored potential differences between murine and human dermal fibroblasts. In principle, primary fibroblasts are highly susceptible to HSV-1; however, we found a delayed infection onset in human compared to murine cells. Intriguingly, only a minor delayed onset of infection was evident in collagen-embedded compared to unembedded human fibroblasts, although expression of the receptor nectin-1 dropped after collagen embedding. This finding is in contrast to previous observations with murine fibroblasts where collagen embedding delayed infection. The application of latex beads revealed limited penetration in the dermis, which was more pronounced in the human than in the murine dermis, supporting the species-specific differences already observed for HSV-1 invasion. Our results suggest that the distinct organization of human and murine dermis contributes to the presence and accessibility of the HSV-1 receptors as well as to the variable barrier function of the extracellular matrix. These contributions, in turn, give rise to inefficient viral access to cells in the dermis while dermal fibroblasts in culture are well infected. IMPORTANCE Dermal fibroblasts are exposed to HSV-1 upon invasion in skin during in vivo infection. Thus, fibroblasts represent a widely used experimental tool to understand virus-host cell interactions and are highly susceptible in culture. The spectrum of fibroblasts' characteristics in their in vivo environment, however, clearly differs from the observations under cell culture conditions, implying putative variations in virus-cell interactions. This becomes evident when ex vivo infection studies in murine as well as human dermis revealed the rather inefficient penetration of HSV-1 in the tissue and uptake in the dermal fibroblasts. Here, we initiated studies to explore the contributions of receptor presence and accessibility to efficient infection of dermal fibroblasts. Our results strengthen the heterogeneity of murine and human dermis and imply that the interplay between dermal barrier function and receptor presence determine how well HSV-1 penetrates the dermis. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
131. Extracellular vimentin is an attachment factor that facilitates SARS-CoV-2 entry into human endothelial cells.
- Author
-
Amraei, Razie, Chaoshuang Xia, Olejnik, Judith, White, Mitchell R., Napoleon, Marc A., Lotfollahzadeh, Saran, Hauser, Blake M., Schmidt, Aaron G., Chitalia, Vipul, Mühlberger, Elke, Costello, Catherine E., and Rahimi, Nader
- Subjects
- *
INTERMEDIATE filament proteins , *ENDOTHELIAL cells , *LIQUID chromatography-mass spectrometry , *VIMENTIN , *SARS-CoV-2 , *COMMERCIAL products - Abstract
SARS-CoV-2 entry into host cells is a crucial step for virus tropism, transmission, and pathogenesis. Angiotensin-converting enzyme 2 (ACE2) has been identified as the primary entry receptor for SARS-CoV-2; however, the possible involvement of other cellular components in the viral entry has not yet been fully elucidated. Here we describe the identification of vimentin (VIM), an intermediate filament protein widely expressed in cells of mesenchymal origin, as an important attachment factor for SARS-CoV-2 on human endothelial cells. Using liquid chromatography-tandem mass spectrometry, we identified VIM as a protein that binds to the SARS-CoV-2 spike (S) protein. We showed that the S-protein receptor binding domain (RBD) is sufficient for S-protein interaction with VIM. Further analysis revealed that extracellular VIM binds to SARS-CoV-2 S-protein and facilitates SARS-CoV-2 infection, as determined by entry assays performed with pseudotyped viruses expressing S and with infectious SARS-CoV-2. Coexpression of VIM with ACE2 increased SARS-CoV-2 entry in HEK-293 cells, and shRNA-mediated knockdown of VIM significantly reduced SARS-CoV-2 infection of human endothelial cells. Moreover, incubation of A549 cells expressing ACE2 with purified VIM increased pseudotyped SARS-CoV-2-S entry. CR3022 antibody, which recognizes a distinct epitope on SARS-CoV-2-S-RBD without interfering with the binding of the spike with ACE2, inhibited the binding of VIM with CoV-2 S-RBD, and neutralized viral entry in human endothelial cells, suggesting a key role for VIM in SARS-CoV-2 infection of endothelial cells. This work provides insight into the pathogenesis of COVID-19 linked to the vascular system, with implications for the development of therapeutics and vaccines. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
132. NPC1-regulated dynamic of clathrin-coated pits is essential for viral entry.
- Author
-
Li, Guoli, Su, Bingqian, Fu, Pengfei, Bai, Yilin, Ding, Guangxu, Li, Dahua, Wang, Jiang, Yang, Guoyu, and Chu, Beibei
- Abstract
Viruses utilize cellular lipids and manipulate host lipid metabolism to ensure their replication and spread. Therefore, the identification of lipids and metabolic pathways that are suitable targets for antiviral development is crucial. Using a library of compounds targeting host lipid metabolic factors and testing them for their ability to block pseudorabies virus (PRV) and vesicular stomatitis virus (VSV) infection, we found that U18666A, a specific inhibitor of Niemann-Pick C1 (NPC1), is highly potent in suppressing the entry of diverse viruses including pseudotyped severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). NPC1 deficiency markedly attenuates viral growth by decreasing cholesterol abundance in the plasma membrane, thereby inhibiting the dynamics of clathrin-coated pits (CCPs), which are indispensable for clathrin-mediated endocytosis. Significantly, exogenous cholesterol can complement the dynamics of CCPs, leading to efficient viral entry and infectivity. Administration of U18666A improves the survival and pathology of PRV- and influenza A virus-infected mice. Thus, our studies demonstrate a unique mechanism by which NPC1 inhibition achieves broad antiviral activity, indicating a potential new therapeutic strategy against SARS-CoV-2, as well as other emerging viruses. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
133. Air-liquid interphase culture confers SARS-CoV-2 susceptibility to A549 alveolar epithelial cells.
- Author
-
Sasaki, Michihito, Kishimoto, Mai, Itakura, Yukari, Tabata, Koshiro, Intaruck, Kittiya, Uemura, Kentaro, Toba, Shinsuke, Sanaki, Takao, Sato, Akihiko, Hall, William W., Orba, Yasuko, and Sawa, Hirofumi
- Subjects
- *
SARS-CoV-2 , *EPITHELIAL cells , *ANGIOTENSIN converting enzyme , *KI-67 antigen , *ANGIOTENSIN II - Abstract
The human lung cell A549 is susceptible to infection with a number of respiratory viruses. However, A549 cells are resistant to Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) infection in conventional submerged culture, and this would appear to be due to low expression levels of the SARS-CoV-2 entry receptor: angiotensin-converting enzyme-2 (ACE2). Here, we examined SARS-CoV-2 susceptibility to A549 cells after adaptation to air-liquid interface (ALI) culture. A549 cells in ALI culture yielded a layer of mucus on their apical surface, exhibited decreased expression levels of the proliferation marker KI-67 and intriguingly became susceptible to SARS-CoV-2 infection. We found that A549 cells increased the endogenous expression levels of ACE2 and TMPRSS2 following adaptation to ALI culture conditions. Camostat, a TMPRSS2 inhibitor, reduced SARS-CoV-2 infection in ALI-cultured A549 cells. These findings indicate that ALI culture switches the phenotype of A549 cells from resistance to susceptibility to SARS-CoV-2 infection through upregulation of ACE2 and TMPRSS2. • A549 cells are resistant to SARS-CoV-2 infection in conventional submerged culture condition. • A549 cells showed susceptibility to SARS-CoV-2 infection in air-liquid interface culture condition. • Air-liquid interface culture upregulated the expression levels of ACE2 and TMPRSS2 in A549 cells. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
134. Kallikreins emerge as new regulators of viral infections.
- Author
-
Pampalakis, Georgios, Zingkou, Eleni, Panagiotidis, Christos, and Sotiropoulou, Georgia
- Subjects
- *
VIRUS diseases , *COVID-19 , *ENZYME activation , *DRUG development , *INFLUENZA , *HEMAGGLUTININ - Abstract
Kallikrein-related peptidases (KLKs) or kallikreins have been linked to diverse (patho) physiological processes, such as the epidermal desquamation and inflammation, seminal clot liquefaction, neurodegeneration, and cancer. Recent mounting evidence suggests that KLKs also represent important regulators of viral infections. It is well-established that certain enveloped viruses, including influenza and coronaviruses, require proteolytic processing of their hemagglutinin or spike proteins, respectively, to infect host cells. Similarly, the capsid protein of the non-enveloped papillomavirus L1 should be proteolytically cleaved for viral uncoating. Consequently, extracellular or membrane-bound proteases of the host cells are instrumental for viral infections and represent potential targets for drug development. Here, we summarize how extracellular proteolysis mediated by the kallikreins is implicated in the process of influenza (and potentially coronavirus and papillomavirus) entry into host cells. Besides direct proteolytic activation of viruses, KLK5 and 12 promote viral entry indirectly through proteolytic cascade events, like the activation of thrombolytic enzymes that also can process hemagglutinin, while additional functions of KLKs in infection cannot be excluded. In the light of recent evidence, KLKs represent potential host targets for the development of new antivirals. Humanized animal models to validate their key functions in viral infections will be valuable. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
135. Ex Vivo Infection of Human Skin with Herpes Simplex Virus 1 Reveals Mechanical Wounds as Insufficient Entry Portals via the Skin Surface.
- Author
-
De La Cruz, Nydia C., Möckel, Maureen, Wirtz, Lisa, Sunaoglu, Katharina, Malter, Wolfram, Zinser, Max, and Knebel-Mörsdorf, Dagmar
- Subjects
- *
HERPES simplex virus , *SKIN infections , *EPIDERMIS , *SKIN , *INFECTION , *WOUNDS & injuries , *SKIN injuries - Abstract
Herpes simplex virus 1 (HSV-1) enters its human host via the skin and mucosa. The open question is how the virus invades this highly protective tissue in vivo to approach its receptors in the epidermis and initiate infection. Here, we performed ex vivo infection studies in human skin to investigate how susceptible the epidermis and dermis are to HSV-1 and whether wounding facilitates viral invasion. Upon ex vivo infection of complete skin, only sample edges with integrity loss demonstrated infected cells. After removal of the dermis, HSV-1 efficiently invaded the basal layer of the epidermis and, from there, gained access to suprabasal layers. This finding supports a high susceptibility of all epidermal layers which correlated with the surface expression of the receptors nectin-1 and herpesvirus entry mediator (HVEM). In contrast, only single infected cells were detected in the separated dermis, where minor expression of the receptors was found. Interestingly, after wounding, nearly no infection of the epidermis was observed via the skin surface. However, if the wounding of the skin samples led to breaks through the dermis, HSV-1 infected mainly keratinocytes via the damaged dermal layer. The application of latex beads revealed only occasional entry via the wounded dermis; however, it facilitated penetration via the wounded skin surface. Thus, we suggest that although the wounded human skin surface allows particle penetration, the skin still provides barriers that prevent HSV-1 from reaching its receptors. IMPORTANCE The human pathogen herpes simplex virus 1 (HSV-1) invades its host via the skin and mucosa, which leads to primary infection of the epithelium. As the various epithelial barriers effectively protect the tissue against viral invasion, successful infection most likely depends on tissue damage. We addressed the initial invasion process in human skin by ex vivo infection to understand how HSV-1 overcomes physical skin barriers and reaches its receptors to enter skin cells. Our results demonstrate that intact skin samples allow viral access only from the edges, while the epidermis is highly susceptible once the basal epidermal layer serves as an initial entry portal. Surprisingly, mechanical wounding did not facilitate HSV-1 entry via the skin surface, although latex beads still penetrated via the lesions. Our results imply that successful invasion of HSV-1 depends on how well the virus can reach its receptors, which was not accomplished by skin lesions under ex vivo conditions. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
136. The Role of L-Selectin in HIV Infection.
- Author
-
Segura, Jason, He, Biao, Ireland, Joanna, Zou, Zhongcheng, Shen, Thomas, Roth, Gwynne, and Sun, Peter D.
- Subjects
HIV infections ,CD4 antigen ,HIV antibodies ,VIRAL proteins ,T cells ,VIRUS-like particles ,VIRAL shedding ,CHEMOKINE receptors - Abstract
HIV envelope glycoprotein is the most heavily glycosylated viral protein complex identified with over 20 glycans on its surface. This glycan canopy is thought to primarily shield the virus from host immune recognition as glycans are poor immunogens in general, however rare HIV neutralizing antibodies nevertheless potently recognize the glycan epitopes. While CD4 and chemokine receptors have been known as viral entry receptor and coreceptor, for many years the role of viral glycans in HIV entry was controversial. Recently, we showed that HIV envelope glycan binds to L-selectin in solution and on CD4 T lymphocytes. The viral glycan and L-selectin interaction functions to facilitate the viral adhesion and entry. Upon entry, infected CD4 T lymphocytes are stimulated to progressively shed L-selectin and suppressing this lectin receptor shedding greatly reduced HIV viral release and caused aggregation of diminutive virus-like particles within experimental infections and from infected primary T lymphocytes derived from both viremic and aviremic individuals. As shedding of L-selectin is mediated by ADAM metalloproteinases downstream of host-cell stimulation, these findings showed a novel mechanism for HIV viral release and offer a potential new class of anti-HIV compounds. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
137. Are Lactobacillus Bulgaricus and Bacillus Calmette-Guérin vaccine suitable for patient protection against SARS-CoV-2 infection?
- Author
-
Avramov, Toma, Vicheva, Dilyana, and Manolova, Antoaneta
- Subjects
- *
SARS-CoV-2 , *COVID-19 , *LACTOBACILLUS , *BCG vaccines , *DRUG approval - Abstract
Before COVID-19 infection caused the global pandemic in 2020, coronavirus diseases were mainly of veterinary interest. This pandemic necessitated the development of protective and therapeutic measures against the spread of SARS-CoV-2. Foods containing representatives of the genus Lactobacillus are an integral part of the daily menu of the Bulgarian people. Our hypothesis is based on studies examining its potential for competitive inhibition of viruses and bacteria by attachment to the surface of enterocytes. Bacillus Calmette-Guérin (BCG) is an integral part of the vaccination calendar in the Republic of Bulgaria. In the literature, many clinical studies show that the administration of BCG vaccine limits the SARS-CoV-2 antigens and, consequently, is able to induce protection for COVID-19, by activating the specific, innate immune system. The lack of definitively approved treatment necessitates finding ways to limit the spread of COVID-19 until final drug approval. We believe that the use of dietary components in the context of competitive inhibition and the vaccination schedule for protection in coronavirus-related diseases is applicable. We hypothesize that Lactobacillus and BCG may play a protective effect against SARS-CoV-2 infection alone or in combination in healthy individuals. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
138. Strategies and Progress in CXCR4-Targeted Anti-Human Immunodeficiency Virus (HIV) Therapeutic Development.
- Author
-
Huang, Lina S M, Snyder, Evan Y, and Schooley, Robert T
- Subjects
- *
HIV infections , *ANTI-HIV agents , *SMALL molecules , *DRUG design , *CELL physiology , *CELL receptors , *CHEMOKINES , *HIV , *PEPTIDES - Abstract
The acquired immunodeficiency syndrome (AIDS), caused by the human immunodeficiency virus (HIV), has been a global public health challenge for several decades. The majority of HIV infection is caused by the human immunodeficiency virus type 1 (HIV-1), which enters and infects a host cell via the cell surface proteins of CD4 as the primary receptor, and chemokine receptors CXCR4 or CCR5 as the coreceptor–then undergoing replication using the cell's intracellular machinery. Whereas many drugs targeting CCR5-mediated entry or HIV-1 replication via reverse transcriptase or proteases have long been used clinically, agents targeting CXCR4 are yet to be advanced to clinical application. Here in this review we highlight some of the strategies for and progress made in the discovery of novel small molecules, peptides, and larger molecules that target CXCR4, and their future prospects for translation into the clinic as a new class of anti-HIV therapeutics. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
139. Molecular basis for higher affinity of SARS‐CoV‐2 spike RBD for human ACE2 receptor.
- Author
-
Delgado, Julián M., Duro, Nalvi, Rogers, David M., Tkatchenko, Alexandre, Pandit, Sagar A., and Varma, Sameer
- Abstract
Severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2) has caused substantially more infections, deaths, and economic disruptions than the 2002‐2003 SARS‐CoV. The key to understanding SARS‐CoV‐2's higher infectivity lies partly in its host receptor recognition mechanism. Experiments show that the human angiotensin converting enzyme 2 (ACE2) protein, which serves as the primary receptor for both CoVs, binds to the receptor binding domain (RBD) of CoV‐2's spike protein stronger than SARS‐CoV's spike RBD. The molecular basis for this difference in binding affinity, however, remains unexplained from X‐ray structures. To go beyond insights gained from X‐ray structures and investigate the role of thermal fluctuations in structure, we employ all‐atom molecular dynamics simulations. Microseconds‐long simulations reveal that while CoV and CoV‐2 spike‐ACE2 interfaces have similar conformational binding modes, CoV‐2 spike interacts with ACE2 via a larger combinatorics of polar contacts, and on average, makes 45% more polar contacts. Correlation analysis and thermodynamic calculations indicate that these differences in the density and dynamics of polar contacts arise from differences in spatial arrangements of interfacial residues, and dynamical coupling between interfacial and non‐interfacial residues. These results recommend that ongoing efforts to design spike‐ACE2 peptide blockers will benefit from incorporating dynamical information as well as allosteric coupling effects. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
140. TMPRSS2 and glycan receptors synergistically facilitate coronavirus entry.
- Author
-
Wang, Haofeng, Liu, Xiaoce, Zhang, Xiang, Zhao, Zhuoqian, Lu, Yuchi, Pu, Dingzhe, Zhang, Zeyang, Chen, Jie, Wang, Yajie, Li, Mengfei, Dong, Xuxue, Duan, Yinkai, He, Yujia, Mao, Qiyu, Guo, Hangtian, Sun, Haoran, Zhou, Yihan, Yang, Qi, Gao, Yan, and Yang, Xiuna
- Subjects
- *
BINDING site assay , *CELLULAR recognition , *CORONAVIRUSES , *MUTAGENESIS , *COVID-19 - Abstract
The entry of coronaviruses is initiated by spike recognition of host cellular receptors, involving proteinaceous and/or glycan receptors. Recently, TMPRSS2 was identified as the proteinaceous receptor for HCoV-HKU1 alongside sialoglycan as a glycan receptor. However, the underlying mechanisms for viral entry remain unknown. Here, we investigated the HCoV-HKU1C spike in the inactive, glycan-activated, and functionally anchored states, revealing that sialoglycan binding induces a conformational change of the NTD and promotes the neighboring RBD of the spike to open for TMPRSS2 recognition, exhibiting a synergistic mechanism for the entry of HCoV-HKU1. The RBD of HCoV-HKU1 features an insertion subdomain that recognizes TMPRSS2 through three previously undiscovered interfaces. Furthermore, structural investigation of HCoV-HKU1A in combination with mutagenesis and binding assays confirms a conserved receptor recognition pattern adopted by HCoV-HKU1. These studies advance our understanding of the complex viral-host interactions during entry, laying the groundwork for developing new therapeutics against coronavirus-associated diseases. [Display omitted] • Structures of the viral spike in complex with different receptors are solved • TMPRSS2 and sialoglycan synergistically facilitate the entry of HCoV-HKU1 • The recognition mode of TMPRSS2 by the HCoV-HKU1 spike is shown • The potential host range of HCoV-HKU1 is suggested Coronavirus entry is initiated by the recognition of proteinaceous and/or glycan receptors. Here, cryo-EM analysis of the HCoV-HKU1 spike in the inactive, glycan-activated, and functionally anchored states demonstrates that TMPRSS2 and sialoglycan synergistically facilitate the entry of HCoV-HKU1. Further insights into the interaction mode between TMPRSS2 and the spike are provided, as well as the potential host range of HCoV-HKU1. These data enhance our understanding of viral-host interaction during entry. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
141. Discovery of a subgenotype of human coronavirus NL63 associated with severe lower respiratory tract infection in China, 2018
- Author
-
Yanqun Wang, Xin Li, Wenkuan Liu, Mian Gan, Lu Zhang, Jin Wang, Zhaoyong Zhang, Airu Zhu, Fang Li, Jing Sun, Guoxian Zhang, Zhen Zhuang, Jiaying Luo, Dehui Chen, Shuyan Qiu, Li Zhang, Duo Xu, Chris Ka Pun Mok, Fuchun Zhang, Jingxian Zhao, Rong Zhou, and Jincun Zhao
- Subjects
human coronavirus NL63 ,new subgenotype ,pneumonia ,whole-genome sequencing ,phylogenetic analysis ,viral entry ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
ABSTRACTHuman coronavirus NL63 (HCoV-NL63) is primarily associated with common cold in children, elderly and immunocompromised individuals. Outbreaks caused by HCoV-NL63 are rare. Here we report a cluster of HCoV-NL63 cases with severe lower respiratory tract infection that arose in Guangzhou, China, in 2018. Twenty-three hospitalized children were confirmed to be HCoV-NL63 positive, and most of whom were hospitalized with severe pneumonia or acute bronchitis. Whole genomes of HCoV-NL63 were obtained using next-generation sequencing. Phylogenetic and single amino acid polymorphism analyses showed that this outbreak was associated with two subgenotypes (C3 and B) of HCoV-NL63. Half of patients were identified to be related to a new subgenotype C3. One unique amino acid mutation at I507 L in spike protein receptor binding domain (RBD) was detected, which segregated this subgenotype C3 from other known subgenotypes. Pseudotyped virus bearing the I507 L mutation in RBD showed enhanced entry into host cells as compared to the prototype virus. This study proved that HCoV-NL63 was undergoing continuous mutation and has the potential to cause severe lower respiratory disease in humans.
- Published
- 2020
- Full Text
- View/download PDF
142. Bat SARS-Like WIV1 coronavirus uses the ACE2 of multiple animal species as receptor and evades IFITM3 restriction via TMPRSS2 activation of membrane fusion
- Author
-
Mei Zheng, Xuesen Zhao, Shuangli Zheng, Danying Chen, Pengcheng Du, Xinglin Li, Dong Jiang, Ju-Tao Guo, Hui Zeng, and Hanxin Lin
- Subjects
SARS-like coronavirus WIV1 ,ACE2 receptor ,viral entry ,IFITM ,TMPRSS2 ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
ABSTRACTDiverse SARS-like coronaviruses (SL-CoVs) have been identified from bats and other animal species. Like SARS-CoV, some bat SL-CoVs, such as WIV1, also use angiotensin converting enzyme 2 (ACE2) from human and bat as entry receptor. However, whether these viruses can also use the ACE2 of other animal species as their receptor remains to be determined. We report herein that WIV1 has a broader tropism to ACE2 orthologs than SARS-CoV isolate Tor2. Among the 9 ACE2 orthologs examined, human ACE2 exhibited the highest efficiency to mediate the infection of WIV1 pseudotyped virus. Our findings thus imply that WIV1 has the potential to infect a wide range of wild animals and may directly jump to humans. We also showed that cell entry of WIV1 could be restricted by interferon-induced transmembrane proteins (IFITMs). However, WIV1 could exploit the airway protease TMPRSS2 to partially evade the IFITM3 restriction. Interestingly, we also found that amphotericin B could enhance the infectious entry of SARS-CoVs and SL-CoVs by evading IFITM3-mediated restriction. Collectively, our findings further underscore the risk of exposure to animal SL-CoVs and highlight the vulnerability of patients who take amphotericin B to infection by SL-CoVs, including the most recently emerging (SARS-CoV-2).
- Published
- 2020
- Full Text
- View/download PDF
143. Targeting the endolysosomal host-SARS-CoV-2 interface by clinically licensed functional inhibitors of acid sphingomyelinase (FIASMA) including the antidepressant fluoxetine
- Author
-
Sebastian Schloer, Linda Brunotte, Jonas Goretzko, Angeles Mecate-Zambrano, Nadia Korthals, Volker Gerke, Stephan Ludwig, and Ursula Rescher
- Subjects
SARS-CoV-2 ,IAV ,FIASMA ,fluoxetine ,viral entry ,endolysosomal interference ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
ABSTRACTThe Coronavirus Disease 2019 (COVID-19) pandemic caused by the Severe Acute Respiratory Syndrome Related Coronavirus 2 (SARS-CoV-2) is a global health emergency. As only very limited therapeutic options are clinically available, there is an urgent need for the rapid development of safe, effective, and globally available pharmaceuticals that inhibit SARS-CoV-2 entry and ameliorate COVID-19 severity. In this study, we explored the use of small compounds acting on the homeostasis of the endolysosomal host-pathogen interface, to fight SARS-CoV-2 infection. We find that fluoxetine, a widely used antidepressant and a functional inhibitor of acid sphingomyelinase (FIASMA), efficiently inhibited the entry and propagation of SARS-CoV-2 in the cell culture model without cytotoxic effects and also exerted potent antiviral activity against two currently circulating influenza A virus subtypes, an effect which was also observed upon treatment with the FIASMAs amiodarone and imipramine. Mechanistically, fluoxetine induced both impaired endolysosomal acidification and the accumulation of cholesterol within the endosomes. As the FIASMA group consists of a large number of small compounds that are well-tolerated and widely used for a broad range of clinical applications, exploring these licensed pharmaceuticals may offer a variety of promising antivirals for host-directed therapy to counteract enveloped viruses, including SARS-CoV-2.
- Published
- 2020
- Full Text
- View/download PDF
144. Ezrin is essential for the entry of Japanese encephalitis virus into the human brain microvascular endothelial cells
- Author
-
Yan-Gang Liu, Yang Chen, Xiaohang Wang, Ping Zhao, Yongzhe Zhu, and Zhongtian Qi
- Subjects
Brain microvascular endothelial cells ,caveolin-1 ,ezrin ,Japanese encephalitis virus ,Src ,viral entry ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
ABSTRACTJapanese encephalitis virus (JEV) remains the predominant cause of viral encephalitis worldwide. It reaches the central nervous system upon crossing the blood–brain barrier through pathogenic mechanisms that are not completely understood. Here, using a high-throughput siRNA screening assay combined with verification experiments, we found that JEV enters the primary human brain microvascular endothelial cells (HBMEC) through a caveolae-mediated endocytic pathway. The role of ezrin, an essential host factor for JEV entry based on our screening, in caveolae-mediated JEV internalization was investigated. We observed that JEV internalization in HBMEC is largely dependent on ezrin-mediated actin cytoskeleton polymerization. Moreover, Src, a protein predicted by a STRING database search, was found to be required in JEV entry. By a variety of pharmacological inhibition and immunoprecipitation assays, we found that Src, ezrin, and caveolin-1 were sequentially activated and formed a complex during JEV infection. A combination of in vitro kinase assay and subcellular analysis demonstrated that ezrin is essential for Src-caveolin-1 interactions. In vivo, both Src and ezrin inhibitors protected ICR suckling mice against JEV-induced mortality and diminished mouse brain viral load. Therefore, JEV entry into HBMEC requires the activation of the Src-ezrin-caveolin-1 signalling axis, which provides potential targets for restricting JEV infection.
- Published
- 2020
- Full Text
- View/download PDF
145. SUMO pathway, blood coagulation and oxidative stress in SARS-CoV-2 infection
- Author
-
Iman Hassan Ibrahim and Doha El-Sayed Ellakwa
- Subjects
SARS CoV-2 ,COVID-19 ,Protein degradation ,Viral entry ,Blood coagulation ,ACE2 and TMPRSS2 ,Biology (General) ,QH301-705.5 ,Biochemistry ,QD415-436 - Abstract
Severe Acute Respiratory Syndrome Corona Virus 2 (SARS CoV-2) is currently an international pandemic causing coronavirus disease 19 (COVID-19). Viral entry requires ACE2 and transmembrane protease serine 2 (TMPRSS2) for membrane fusion or through endosomal pathway. This Study aims to assess transcriptomic changes and differentially expressed genes (DFGs) in COVID-19. Methods: Transcriptomic data of the publicly available dataset (GSE147507) was quantile normalized and analysed for DFGs, network analysis and pathway analysis. Results: DFG sets showed that 8 genes (SAE1, AEBP2, ATP1A1, DKK3, MAFF, NUDC, TRAP1, and VAV1) were significantly dysregulated in all studied groups. Functional analysis revealed that negative regulation of glucocorticoid biosynthesis, protein SUMOylation (SAE1), blood coagulation (VAV1) and cellular response to stress were affected by SARS CoV-2 infection. Cell line transduction with ACE2 vector didn't show significant changes in the dysregulated pathways. Also, no significant change was observed in expression levels of ACE2 or TMPRSS2 in response to SARS CoV-2 infection. Further analysis showed dysregulation of several genes in the SUMOylation pathway and blood coagulation process in human and cell lines transcriptome. Also, several Cathepsins proteases were significantly dysregulated in case of SARS CoV-2 infection. Genes related to cellular response to stress such as TRAP-1 and NOX were dysregulated in cases of SARS CoV-2 infection. Conclusion: Dysregulation in genes of protein SUMOylation, blood coagulation and response to oxidative stress pathways in SARS CoV-2 infection could be critical for disease progression. Drugs acting on SUMO pathway, VAV1, NOX genes could be studied for potential benefit to COVID-19 patients.
- Published
- 2021
- Full Text
- View/download PDF
146. Heat Shock Protein 70 (HSP70) Family in Dengue Virus Infection
- Author
-
Kanlaya, Rattiyaporn, Thongboonkerd, Visith, Asea, Alexzander A. A., Series Editor, Calderwood, Stuart K., Series Editor, Asea, Alexzander A A, editor, and Kaur, Punit, editor
- Published
- 2018
- Full Text
- View/download PDF
147. Exploring peptide studies related to SARS-CoV to accelerate the development of novel therapeutic and prophylactic solutions against COVID-19.
- Author
-
Madhavan, Maya, AlOmair, Lamya A., KS, Deepthi, and Mustafa, Sabeena
- Abstract
Recent advances in peptide research revolutionized therapeutic discoveries for various infectious diseases. In view of the ongoing threat of the COVID-19 pandemic, there is an urgent need to develop potential therapeutic options. Intense and accomplishing research is being carried out to develop broad-spectrum vaccines and treatment options for corona viruses, due to the risk of recurrent infection by the existing strains or pandemic outbreaks by new mutant strains. Developing a novel medicine is costly and time consuming, which increases the value of repurposing existing therapies. Since, SARS-CoV-2 shares significant genomic homology with SARS-CoV, we have summarized various peptides identified against SARS-CoV using in silico and molecular studies and also the peptides effective against SARS-CoV-2. Dissecting the molecular mechanisms underlying viral infection could yield fundamental insights in the discovery of new antiviral agents, targeting viral proteins or host factors. We postulate that these peptides can serve as effective components for therapeutic options against SARS-CoV-2, supporting clinical scientists globally in selectively identifying and testing the therapeutic and prophylactic agents for COVID-19 treatment. In addition, we also summarized the latest updates on peptide therapeutics against SARS-CoV-2. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
148. A systems‐level study reveals host‐targeted repurposable drugs against SARS‐CoV‐2 infection.
- Author
-
Chen, Fangyuan, Shi, Qingya, Pei, Fen, Vogt, Andreas, Porritt, Rebecca A, Garcia, Gustavo, Gomez, Angela C, Cheng, Mary Hongying, Schurdak, Mark E, Liu, Bing, Chan, Stephen Y, Arumugaswami, Vaithilingaraja, Stern, Andrew M, Taylor, D Lansing, Arditi, Moshe, and Bahar, Ivet
- Subjects
SARS-CoV-2 ,COVID-19 ,INVESTIGATIONAL drugs ,SMALL molecules ,CELL fusion ,ANTIVIRAL agents - Abstract
Understanding the mechanism of SARS‐CoV‐2 infection and identifying potential therapeutics are global imperatives. Using a quantitative systems pharmacology approach, we identified a set of repurposable and investigational drugs as potential therapeutics against COVID‐19. These were deduced from the gene expression signature of SARS‐CoV‐2‐infected A549 cells screened against Connectivity Map and prioritized by network proximity analysis with respect to disease modules in the viral–host interactome. We also identified immuno‐modulating compounds aiming at suppressing hyperinflammatory responses in severe COVID‐19 patients, based on the transcriptome of ACE2‐overexpressing A549 cells. Experiments with Vero‐E6 cells infected by SARS‐CoV‐2, as well as independent syncytia formation assays for probing ACE2/SARS‐CoV‐2 spike protein‐mediated cell fusion using HEK293T and Calu‐3 cells, showed that several predicted compounds had inhibitory activities. Among them, salmeterol, rottlerin, and mTOR inhibitors exhibited antiviral activities in Vero‐E6 cells; imipramine, linsitinib, hexylresorcinol, ezetimibe, and brompheniramine impaired viral entry. These novel findings provide new paths for broadening the repertoire of compounds pursued as therapeutics against COVID‐19. SYNOPSIS: A systems pharmacology approach is proposed to identify small molecules and drugs that target host cells and have anti‐SARS‐CoV‐2 effects. Shortlisted drug candidates are experimentally validated. Host‐targeted antiviral and anti‐cytokine compounds are inferred from the transcriptome data of SARS‐CoV‐2‐infected cells.Salmeterol exhibits a strong antiviral effect in SARS‐CoV‐2‐infected Vero‐E6 cells.Syncytia formation assays confirm that linsitinib is a potent inhibitor of viral entry.Additional compounds are proposed for adjuvant anti‐inflammatory therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
149. Host Factors and Pathways Involved in the Entry of Mosquito-Borne Alphaviruses.
- Author
-
De Caluwé, Lien, Ariën, Kevin K., and Bartholomeeusen, Koen
- Subjects
- *
ALPHAVIRUSES , *CHIKUNGUNYA virus , *ARBOVIRUS diseases , *ARBOVIRUSES , *ALPHAVIRUS diseases , *TOGAVIRUSES , *MOSQUITOES , *ENDOCYTOSIS , *TROPISMS - Abstract
Chikungunya virus (CHIKV) is an arthropod-borne virus that has re-emerged recently and has spread to previously unaffected regions, resulting in millions of infections worldwide. The genus Alphavirus , in the family Togaviridae, contains several members with a similar potential for epidemic emergence. In order for CHIKV to replicate in targeted cell types it is essential for the virus to enter these cells. In this review, we summarize our current understanding of the versatile and promiscuous steps in CHIKV binding and entry into human and mosquito host cells. We describe the different entry pathways, receptors, and attachment factors so far described for CHIKV and other mosquito-borne alphaviruses and discuss them in the context of tissue tropism and potential therapeutic targeting. In the replication cycle of alphaviruses, the entry step is the first essential step to allow efficient production of new viral particles. The main entry pathway of alphaviruses is clathrin-mediated endocytosis. Fusion requires a low pH and takes place in the endosome. However, other, clathrin-independent, pathways that facilitate alphavirus entry have also been described. Some host factors that can mediate alphavirus entry have been identified but entry in the absence of these host factors is still possible. Alphavirus entry appears to be a promiscuous process following multiple routes where various pathways, attachment factors, and receptors can be used, assuring a broad tissue tropism. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
150. Spiking dependence of SARS‐CoV‐2 pathogenicity on TMPRSS2.
- Author
-
Abbasi, Asim Z., Kiyani, Dania A., Hamid, Syeda M., Saalim, Muhammad, Fahim, Ammad, and Jalal, Nasir
- Subjects
SARS-CoV-2 ,COVID-19 ,COVID-19 treatment ,VIRAL proteins ,ANDROGEN receptors ,VIRAL envelope proteins - Abstract
Epidemiological data shows a discrepancy in COVID‐19 susceptibility and outcomes with some regions being more heavily affected than others. However, the factors that determine host susceptibility and pathogenicity remain elusive. An increasing number of publications highlight the role of Transmembrane Serine Protease 2 (TMPRSS2) in the susceptibility of the host cell to SARS‐CoV‐2. Cleavage of viral spike protein via the host cell's TMPRSS2 enzyme activity mediates viral entry into the host cell. The enzyme synthesis is regulated by the TMPRSS2 gene, which has also been implicated in the entry mechanisms of previously reported Coronavirus infections. In this review, we have investigated the pathogenicity of SARS‐CoV‐2 and disease susceptibility dependence on the TMPRSS2 gene as expressed in various population groups. We further discuss how the differential expression of this gene in various ethnic groups can affect the SARS‐CoV‐2 infection and Coronavirus disease (COVID)‐19 outcomes. Moreover, promising new TMPRSS2 protease blockers and inhibitors are discussed for COVID‐19 treatment. Highlights: 1. Entry of SARS‐CoV‐2 into a host cell depends on host protease TMPRSS2. 2. TMPRSS2 gene has localized expression throughout the human body but highly expressed in cells of the respiratory tract (primary target of SARS‐CoV‐2 in humans), gastrointestinal tract, kidneys and prostate. 3. Differences in expression of TMPRSS2 gene in the respiratory among different population groups can be a basis for discrepancy observed in COVID‐19 susceptibility and disease outcomes. 4. Drugs based on the inhibition or blockage of TMPRSS2 protease are undergoing clinical trials as a therapeutic option. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.