Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell Mol Immunol ; 19(6): 702-714, 2022 06.
Article in English | MEDLINE | ID: mdl-35332300

ABSTRACT

Innate immunity plays critical antiviral roles. The highly virulent avian influenza viruses (AIVs) H5N1, H7N9, and H5N6 can better escape host innate immune responses than the less virulent seasonal H1N1 virus. Here, we report a mechanism by which transcriptional readthrough (TRT)-mediated suppression of innate immunity occurs post AIV infection. By using cell lines, mouse lungs, and patient PBMCs, we showed that genes on the complementary strand ("trans" genes) influenced by TRT were involved in the disruption of host antiviral responses during AIV infection. The trans-TRT enhanced viral lethality, and TRT abolishment increased cell viability and STAT1/2 expression. The viral NS1 protein directly bound to SSU72, and degradation of SSU72 induced TRT. SSU72 overexpression reduced TRT and alleviated mouse lung injury. Our results suggest that AIVs infection induce TRT by reducing SSU72 expression, thereby impairing host immune responses, a molecular mechanism acting through the NS1-SSU72-trans-TRT-STAT1/2 axis. Thus, restoration of SSU72 expression might be a potential strategy for preventing AIV pandemics.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A Virus, H5N1 Subtype , Influenza A Virus, H7N9 Subtype , Influenza, Human , Animals , Antiviral Agents , Humans , Immunity, Innate , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/metabolism , Influenza A Virus, H7N9 Subtype/metabolism , Mice , Phosphoprotein Phosphatases , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
2.
Cancer Res ; 75(19): 4198-210, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26359459

ABSTRACT

Platelet-activating factor receptor (PAFR), a G-protein-coupled receptor, has been implicated in tumorigenesis, but its contributions to metastatic progression have not been investigated. Here, we show that PAFR is overexpressed in non-small cell lung cancer (NSCLC) as well as in breast, colorectal, and gastric carcinomas. Expression of PAFR correlates closely with clinical stages, survival time, and distant metastasis. In human NSCLC cells, activation of the PAF/PAFR signaling axis accentuated malignant character, including by stimulating epithelial-mesenchymal transition (EMT). In contrast, silencing PAFR in aggressive NSCLC cells inhibited these effects. Mechanistic investigations showed that PAFR stimulated EMT by activating STAT3 via upregulation of G-protein-dependent SRC or JAK2 kinase activity. Notably, STAT3 transcriptionally elevated PAFR expression. Thus, activation of PAFR in NSCLC cells initiated a forward feedback loop responsible for mediating the aggressive malignant character of NSCLC cells in vitro and in vivo. Reinforcing this reciprocal activation loop, PAF/PAFR signaling also upregulated IL6 expression and thereby STAT3 activation. Overall, our results elucidated an important role for PAFR dysregulation in the pathogenicity of NSCLC and unraveled a forward feedback loop between PAFR and STAT3 that acts to drive the malignant progression of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Epithelial-Mesenchymal Transition/physiology , Lung Neoplasms/pathology , Neoplasm Proteins/physiology , Platelet Membrane Glycoproteins/physiology , Receptors, G-Protein-Coupled/physiology , STAT3 Transcription Factor/physiology , Adult , Aged , Animals , Breast Neoplasms/metabolism , Carcinoma/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Disease Progression , Enzyme Activation , Feedback, Physiological , Female , GTP-Binding Proteins/physiology , Heterografts , Humans , Interleukin-6/biosynthesis , Interleukin-6/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Transplantation , Protein Kinases/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction , Stomach Neoplasms/metabolism , Up-Regulation
3.
PLoS One ; 10(8): e0136613, 2015.
Article in English | MEDLINE | ID: mdl-26317987

ABSTRACT

B cell hybridomas are an important source of monoclonal antibodies. In this paper, we developed a high-throughput method to characterize mouse IgG antibodies using surface plasmon resonance technology. This assay rapidly determines their sub-isotypes, whether they bind native antigen and their approximate affinities for the antigen using only 50 µl of hybridoma cell culture supernatant. Moreover, we found that mouse hybridomas secreting IgG antibodies also have membrane form IgG expression without Igα. Based on this surface IgG, we used flow cytometry to isolate rare γ2a isotype switched variants from a γ2b antibody secreting hybridoma cell line. Also, we used fluorescent antigen to single cell sort antigen binding hybridoma cells from bulk mixture of fused hybridoma cells instead of the traditional multi-microwell plate screening and limiting dilution sub-cloning thus saving time and labor. The IgG monoclonal antibodies specific for the native antigen identified with these methods are suitable for in vivo therapeutic uses, but also for sandwich ELISA assays, histology, flow cytometry, immune precipitation and x-ray crystallography.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Antigens/chemistry , B-Lymphocytes/chemistry , Hybridomas/chemistry , Immunoglobulin G/chemistry , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Antigens/immunology , B-Lymphocytes/immunology , Hybridomas/immunology , Immunoglobulin G/immunology , Mice
4.
J Virol ; 89(20): 10347-58, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26246576

ABSTRACT

UNLABELLED: As a recycling center, lysosomes are filled with numerous acid hydrolase enzymes that break down waste materials and invading pathogens. Recently, lysosomal cell death has been defined as "lysosomal membrane permeabilization and the consequent leakage of lysosome contents into cytosol." Here, we show that the neuraminidase (NA) of H5N1 influenza A virus markedly deglycosylates and degrades lysosome-associated membrane proteins (LAMPs; the most abundant membrane proteins of lysosome), which induces lysosomal rupture, and finally leads to cell death of alveolar epithelial carcinoma A549 cells and human tracheal epithelial cells. The NA inhibitors peramivir and zanamivir could effectively block the deglycosylation of LAMPs, inhibit the virus cell entry, and prevent cell death induced by the H5N1 influenza virus. The NA of seasonal H1N1 virus, however, does not share these characteristics. Our findings not only reveal a novel role of NA in the early stage of the H5N1 influenza virus life cycle but also elucidate the molecular mechanism of lysosomal rupture crucial for influenza virus induced cell death. IMPORTANCE: The integrity of lysosomes is vital for maintaining cell homeostasis, cellular defense and clearance of invading pathogens. This study shows that the H5N1 influenza virus could induce lysosomal rupture through deglycosylating lysosome-associated membrane proteins (LAMPs) mediated by the neuraminidase activity of NA protein. NA inhibitors such as peramivir and zanamivir could inhibit the deglycosylation of LAMPs and protect lysosomes, which also further interferes with the H5N1 influenza virus infection at early stage of life cycle. This work is significant because it presents new concepts for NA's function, as well as for influenza inhibitors' mechanism of action, and could partially explain the high mortality and high viral load after H5N1 virus infection in human beings and why NA inhibitors have more potent therapeutic effects for lethal avian influenza virus infections at early stage.


Subject(s)
Cell Membrane/enzymology , Lysosomal Membrane Proteins/metabolism , Lysosomes/enzymology , Neuraminidase/metabolism , Viral Proteins/metabolism , Acids, Carbocyclic , Cell Death/drug effects , Cell Line, Tumor , Cell Membrane/chemistry , Cyclopentanes/pharmacology , Cytosol/drug effects , Cytosol/enzymology , Cytosol/virology , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/virology , Guanidines/pharmacology , Humans , Hydrolysis , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/enzymology , Lysosomal Membrane Proteins/chemistry , Lysosomes/drug effects , Lysosomes/virology , Protein Binding , Proteolysis , Respiratory Mucosa/drug effects , Respiratory Mucosa/virology , Species Specificity , Virus Internalization/drug effects , Zanamivir/pharmacology
5.
Arch Biochem Biophys ; 575: 38-45, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25843419

ABSTRACT

The clinical efficacy of cisplatin in esophageal squamous cell carcinoma (ESCC) treatment remains undesirable. Src, a non-receptor tyrosine kinase involved in multiple fields of tumorigenesis, recently has been indicated as a promising therapeutic target in the treatment of solid tumors including ESCC. However, whether inhibition of Src activity can increase cisplatin efficacy in ESCC cells remains unknown. The present study found that inhibition of Src by its inhibitor-dasatinib sensitized ESCC cells to cisplatin in vitro. Our data also suggest a likely mechanism for this synergy that dasatinib reduces expression of critical oncogenic members of the signaling pathways, such as AKT or Stat3, and cisplatin-resistant molecules, such as ERCC1 and BRCA1, under the control of Src. Furthermore, dasatinib could sensitize ESCC cells to another platin-based agent, carboplatin. Therefore, this study provides a potential target for improving cisplatin efficacy in ESCC therapy.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cisplatin/pharmacology , Esophageal Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrimidines/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Thiazoles/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Dasatinib , Drug Synergism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism
6.
Part Fibre Toxicol ; 12: 4, 2015 Mar 07.
Article in English | MEDLINE | ID: mdl-25890286

ABSTRACT

BACKGROUND: Nanoparticles have become a key technology in multiple industries. However, there are growing reports of the toxicity of nanomaterials to humans. In particular, nanomaterials have been linked to lung diseases. The molecular mechanisms of nanoparticle toxicity are largely unexplored. METHODS: Acute lung injury was induced in wild-type mice and angiotensin-coverting enzyme 2 (ACE2) knockout mice by the intratracheal instillation of cationic polyamidoamine dendrimer (PAMAM) nanoparticles. For rescue experiments, losartan (15 mg/kg in PBS) was injected intraperitoneally 30 min before nanoparticle administration. RESULTS: Some PAMAM nanoparticles, but not anionic PAMAM nanoparticles or carbon nanotubes, triggered acute lung failure in mice. Mechanistically, cationic nanoparticles can directly bind ACE2, decrease its activity and down-regulate its expression level in lung tissue, resulting in deregulation of the renin-angiotensin system. Gene inactivation of Ace2 can exacerbate lung injury. Importantly, the administration of losartan, which is an angiotensin II type I receptor antagonist, can ameliorate PAMAM nanoparticle-induced lung injury. CONCLUSIONS: Our data provide molecular insight into PAMAM nanoparticle-induced lung injury and suggest potential therapeutic and screening strategies to address the safety of nanomaterials.


Subject(s)
Acute Lung Injury/chemically induced , Dendrimers/toxicity , Nanoparticles/toxicity , Peptidyl-Dipeptidase A/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Angiotensin-Converting Enzyme 2 , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Cations , Dendrimers/administration & dosage , Dendrimers/chemistry , Down-Regulation , Instillation, Drug , Losartan/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Peptidyl-Dipeptidase A/genetics , Protein Binding , Survival Analysis
7.
Sci China Life Sci ; 57(10): 959-72, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25218824

ABSTRACT

Ebola virus (EBOV) causes a highly lethal hemorrhagic fever syndrome in humans and has been associated with mortality rates of up to 91% in Zaire, the most lethal strain. Though the viral envelope glycoprotein (GP) mediates widespread inflammation and cellular damage, these changes have mainly focused on alterations at the protein level, the role of microRNAs (miRNAs) in the molecular pathogenesis underlying this lethal disease is not fully understood. Here, we report that the mi-RNAs hsa-miR-1246, hsa-miR-320a and hsa-miR-196b-5p were induced in human umbilical vein endothelial cells (HUVECs) following expression of EBOV GP. Among the proteins encoded by predicted targets of these miRNAs, the adhesion-related molecules tissue factor pathway inhibitor (TFPI), dystroglycan1 (DAG1) and the caspase 8 and FADD-like apoptosis regulator (CFLAR) were significantly downregulated in EBOV GP-expressing HUVECs. Moreover, inhibition of hsa-miR-1246, hsa-miR-320a and hsa-miR-196b-5p, or overexpression of TFPI, DAG1 and CFLAR rescued the cell viability that was induced by EBOV GP. Our results provide a novel molecular basis for EBOV pathogenesis and may contribute to the development of strategies to protect against future EBOV pandemics.


Subject(s)
Hemorrhagic Fever, Ebola/genetics , MicroRNAs/genetics , Viral Envelope Proteins/metabolism , Blotting, Western , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/metabolism , Cell Survival , Dystroglycans/metabolism , Ebolavirus , Gene Expression Regulation , HEK293 Cells , High-Throughput Nucleotide Sequencing , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Inflammation , Lipoproteins/metabolism , MicroRNAs/physiology
8.
Sci Signal ; 5(212): ra16, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22355189

ABSTRACT

The threat of a new influenza pandemic has existed since 1997, when the highly pathogenic H5N1 strain of avian influenza A virus infected humans in Hong Kong and spread across Asia, where it continued to infect poultry and people. The human mortality rate of H5N1 infection is about 60%, whereas that of seasonal H1N1 infection is less than 0.1%. The high mortality rate associated with H5N1 infection is predominantly a result of respiratory failure caused by acute lung injury; however, how viral infection contributes to this disease pathology is unclear. Here, we used electron microscopy to show the accumulation of autophagosomes in H5N1-infected lungs from a human cadaver and mice, as well as in infected A549 human epithelial lung cells. We also showed that H5N1, but not seasonal H1N1, induced autophagic cell death in alveolar epithelial cells through a pathway involving the kinase Akt, the tumor suppressor protein TSC2, and the mammalian target of rapamycin. Additionally, we suggest that the hemagglutinin protein of H5N1 may be responsible for stimulating autophagy. When applied prophylactically, reagents that blocked virus-induced autophagic signaling substantially increased the survival rate of mice and substantially ameliorated the acute lung injury and mortality caused by H5N1 infection. We conclude that the autophagic cell death of alveolar epithelial cells likely plays a crucial role in the high mortality rate of H5N1 infection, and we suggest that autophagy-blocking agents might be useful as prophylactics and therapeutics against infection of humans by the H5N1 virus.


Subject(s)
Autophagy/physiology , Influenza A Virus, H5N1 Subtype , Influenza, Human/pathology , Lung/ultrastructure , Signal Transduction/physiology , Analysis of Variance , Animals , Autophagy/drug effects , Autophagy-Related Protein 5 , Blotting, Western , Cell Line , DNA Primers/genetics , Epithelial Cells/physiology , Gene Knockdown Techniques , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Humans , Lung/virology , Mice , Mice, Inbred BALB C , Microscopy, Electron , Microtubule-Associated Proteins/genetics , Oncogene Protein v-akt/metabolism , Phagosomes/pathology , Real-Time Polymerase Chain Reaction , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
9.
PLoS One ; 6(8): e23710, 2011.
Article in English | MEDLINE | ID: mdl-21887302

ABSTRACT

It has been reported that lactoferrin (LF) participates in the host immune response against Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) invasion by enhancing NK cell activity and stimulating neutrophil aggregation and adhesion. We further investigated the role of LF in the entry of SARS pseudovirus into HEK293E/ACE2-Myc cells. Our results reveal that LF inhibits SARS pseudovirus infection in a dose-dependent manner. Further analysis suggested that LF was able to block the binding of spike protein to host cells at 4°C, indicating that LF exerted its inhibitory function at the viral attachment stage. However, LF did not disrupt the interaction of spike protein with angiotensin-converting enzyme 2 (ACE2), the functional receptor of SARS-CoV. Previous studies have shown that LF colocalizes with the widely distributed cell-surface heparan sulfate proteoglycans (HSPGs). Our experiments have also confirmed this conclusion. Treatment of the cells with heparinase or exogenous heparin prevented binding of spike protein to host cells and inhibited SARS pseudovirus infection, demonstrating that HSPGs provide the binding sites for SARS-CoV invasion at the early attachment phase. Taken together, our results suggest that, in addition to ACE2, HSPGs are essential cell-surface molecules involved in SARS-CoV cell entry. LF may play a protective role in host defense against SARS-CoV infection through binding to HSPGs and blocking the preliminary interaction between SARS-CoV and host cells. Our findings may provide further understanding of SARS-CoV pathogenesis and aid in treatment of this deadly disease.


Subject(s)
Heparan Sulfate Proteoglycans/metabolism , Lactoferrin/pharmacology , Severe Acute Respiratory Syndrome/prevention & control , Severe acute respiratory syndrome-related coronavirus/physiology , Virus Internalization , Cell Line , Dose-Response Relationship, Drug , Humans , Lactoferrin/immunology , Lactoferrin/metabolism , Protein Binding/immunology , Severe Acute Respiratory Syndrome/etiology , Severe Acute Respiratory Syndrome/immunology , Virus Internalization/drug effects
10.
Chin Med J (Engl) ; 123(8): 1073-7, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20497717

ABSTRACT

BACKGROUND: Numerous Asian cases of avian influenza virus infection, especially the highly pathogenic strain H5N1, in humans have raised the concern that another influenza pandemic is close. However, there are no effective therapeutic drugs or preventative vaccines available. Hemagglutinin is the membrane glycoprotein of avian influenza virus responsible for receptor binding to human cells and the main immunogenic protein that elicits a strong immune response. Although this protein is of great importance to the study of pathogenesis and vaccine development, its expression and purification are difficult due to high levels of glycosylation. METHODS: In this study, we expressed codon-optimized, full-length hemagglutinin 5 (H5) protein fused with a human IgG Fc tag (H5-Fc) in HEK293 cells. To enhance secretion of this protein, we also deleted the transmembrane domain and the intracellular domain of the H5 protein (H5DeltaTM-Fc). Purified proteins were obtained using a protein A column. RESULTS: ELISA revealed that the yield of soluble H5DeltaTM-Fc protein in the supernatant was about 20 mg/L. Western blotting and fluorescence activated cell sorter (FACS) indicated that the purified H5 protein was correctly folded and biologically active. CONCLUSION: Purification of H5 proteins from mammalian cells could be used for large-scale production of recombinant H5 protein for basic scientific research or the development of vaccines.


Subject(s)
Codon/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Recombinant Proteins/metabolism , Cell Line , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Protein Folding , Recombinant Proteins/genetics
11.
Virus Res ; 136(1-2): 8-15, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18554741

ABSTRACT

Cell entry of severe acute respiratory syndrome coronavirus (SARS-CoV) is mediated by the viral spike (S) protein. Amino acids 319-510 on the S protein have been mapped as the receptor-binding domain (RBD), which mediates binding to the SARS-CoV receptor angiotensin converting enzyme 2 (ACE2) on SARS-CoV susceptible cells. In this study, we expressed a fusion protein containing the human codon-optimized RBD of the SARS-CoV spike protein linked to the Fc portion of human IgG1 (named RBD-Fc) in HEK293 cells. The RBD-Fc protein was purified by affinity chromatography. The flow cytometry assay showed that the purified RBD-Fc protein could bind to ACE2. We demonstrated that the RBD spike protein alone could be internalized into SARS-CoV susceptible cells together with ACE2. We also showed that the removal of N-glycans from the RBD spike protein did not abolish this phenomenon. Our discoveries may have some implications for the development of the SARS vaccine.


Subject(s)
Endocytosis , Membrane Glycoproteins/metabolism , Peptidyl-Dipeptidase A/metabolism , Receptors, Virus/metabolism , Severe acute respiratory syndrome-related coronavirus/physiology , Viral Envelope Proteins/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Cell Line , Chlorocebus aethiops , Flow Cytometry , Humans , Immunoglobulin Fc Fragments/genetics , Membrane Glycoproteins/genetics , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics
12.
Cell Res ; 18(2): 290-301, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18227861

ABSTRACT

While severe acute respiratory syndrome coronavirus (SARS-CoV) was initially thought to enter cells through direct fusion with the plasma membrane, more recent evidence suggests that virus entry may also involve endocytosis. We have found that SARS-CoV enters cells via pH- and receptor-dependent endocytosis. Treatment of cells with either SARS-CoV spike protein or spike-bearing pseudoviruses resulted in the translocation of angiotensin-converting enzyme 2 (ACE2), the functional receptor of SARS-CoV, from the cell surface to endosomes. In addition, the spike-bearing pseudoviruses and early endosome antigen 1 were found to colocalize in endosomes. Further analyses using specific endocytic pathway inhibitors and dominant-negative Eps15 as well as caveolin-1 colocalization study suggested that virus entry was mediated by a clathrin- and caveolae-independent mechanism. Moreover, cholesterol- and sphingolipid-rich lipid raft microdomains in the plasma membrane, which have been shown to act as platforms for many physiological signaling pathways, were shown to be involved in virus entry. Endocytic entry of SARS-CoV may expand the cellular range of SARS-CoV infection, and our findings here contribute to the understanding of SARS-CoV pathogenesis, providing new information for anti-viral drug research.


Subject(s)
Endocytosis , Endosomes/metabolism , Membrane Glycoproteins/metabolism , Peptidyl-Dipeptidase A/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Viral Envelope Proteins/metabolism , Virus Internalization , Adaptor Proteins, Signal Transducing , Angiotensin-Converting Enzyme 2 , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Caveolae/metabolism , Caveolae/virology , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line , Clathrin/genetics , Clathrin/metabolism , Endocytosis/genetics , Endosomes/virology , Humans , Hydrogen-Ion Concentration , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Glycoproteins/genetics , Peptidyl-Dipeptidase A/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Transport , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Severe Acute Respiratory Syndrome/drug therapy , Severe Acute Respiratory Syndrome/genetics , Severe Acute Respiratory Syndrome/metabolism , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...