• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 1
  • Tagged with
  • 6
  • 6
  • 6
  • 5
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • 3
  • 2
  • 2
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

The Membrane Integration of the Hemagglutinin-Neuraminidase Glycoprotein of Newcastle Disease Virus: A Thesis

Wilson, Cheryl Anne 01 May 1989 (has links)
The hemagglutinin-neuraminidase (HN) molecule of Newcastle disease virus (NDV) is an integral membrane glycoprotein that is oriented with its N-terminus in the cytoplasm and its C-terminus external to the infected cell. Single spanning membrane proteins with this type of topology (N-terminus in, C-terminus out) have been classified as Type II glycoproteins, in contrast to the more common Type I glycoproteins, which are oriented in the opposite direction. (C-terminus in, N-terminus out). The membrane integration of HN protein was investigated using a wheat germ translation system to synthesize and integrate HN protein into microsomal membranes in vitro. The insertion and translocation of HN protein into microsomal vesicles was found to occur cotranslationally without signal sequence cleavage. The membrane targeting required both signal recognition particle (SRP) and SRP receptor. Membrane binding assays utilizing HN nascent chain/ribosome/SRP complexes demonstrated that the membrane insertion of HN polypeptide required the presence of GTP, in a way similar to that described for secretory, multispanning and Type I proteins. To investigate further the membrane translocation process of HN protein, the amino terminal region of HN was mutated to determine the role of this region in the membrane integration of HN. The cDNA sequence encoding the bulk of the cytoplasmic tail of the HN glycoprotein was deleted. When transcripts produced from the mutated cDNA were translated in wheat germ extract in the presence of membranes, several abnormalities were identified in the interaction of the mutant protein with membranes. Although translocation and glycosylation of the mutant protein was detected, the efficiency of membrane translocation and the stability of the mutant protein's membrane interaction were reduced. Even though a large proportion of the mutant products remained nontranslocated and unglycosylated, many of these products were inserted into membrane vesicles in a reverse orientation from the wild type HN protein. The aberrant insertion of the mutant protein required both SRP and SRP receptor. Ribosome-bound mutant nascent chains were able to insert into membranes without the addition of GTP or SRP, but this GTP-independent insertion was in reverse. Therefore, the cytoplasmic tail of the HN glycoprotein appears to playa critical role in the maintanence of faithful directionality of the protein's membrane insertion.
2

Newcastle Disease Virus Virulence: Mechanism of the Interferon Antagonistic Activity of the V Protein and Characterization of a Putative Virulence-Specific Antibody to the Attachment Protein: a dissertation

Alamares, Judith G. 05 May 2008 (has links)
Newcastle disease virus (NDV) is a member of the genus Avulavirus of the Paramyxoviridaefamily of enveloped negative-stranded RNA viruses. The virus causes respiratory, neurological, or enteric disease in many species of birds, resulting in significant losses to the poultry industry worldwide. Strains of the virus are classified into three pathotypes based on the severity of disease in chickens. Avirulent strains that produce mild or asymptomatic infections are termed lentogenic, whereas virulent strains are termed velogenic. Strains of intermediate virulence are termed mesogenic. The envelope of NDV virions contains two types of glycoproteins, the hemagglutinin-neuraminidase (HN) and fusion (F) proteins. HN mediates three functions: 1) virus attachment to sialic acid-containing receptors; 2) neuraminidase activity that cleaves sialic acid from progeny virions to prevent self-aggregation; and, 3) complementation of the F protein in the promotion of fusion. Though it is widely accepted that cleavage of a fusion protein precursor is the primary determinant of NDV virulence, it is not the sole determinant. At least two other proteins, HN and the V protein, contribute to virulence. The V protein possesses interferon (IFN) antagonistic activity. The long-range goal of these studies is to understand the roles of HN and V in the differential virulence patterns exhibited by members of the NDV serotype. The first aim is to compare the IFN antagonistic activity of the V protein from a lentogenic and a mesogenic strain of the virus. The results of this study demonstrate that the V protein of the mesogenic strain Beaudette C (BC) exhibits greater IFN antagonistic activity than that of the lentogenic strain La Sota. Hence, the IFN antagonistic activities of the two V proteins correlate with their known virulence properties. Comparison of the C-terminal regions of La Sota and BC V proteins revealed four amino acid differences. The results demonstrate that the IFN antagonistic activity of La Sota V increases when any one of these residues is mutated to the corresponding residue in BC V. Conversely, the IFN antagonistic activity of BC V decreases when any one of these four residues is mutated to the corresponding residue in La Sota V. However, no single residue accounts for the difference in IFN antagonistic activity between the two V proteins. Also, analysis of La Sota V and BC V proteins with multiple mutations in these positions revealed that the four residues are collectively responsible for the difference in the IFN antagonistic activity of the two V proteins. Finally, characterization of chimeric La Sota/BC V proteins showed that the N-terminal region also contributes to the IFN antagonistic activity of V. Contrary to an earlier report, results described here demonstrate that the NDV V protein does not target STAT1 for degradation. However, both La Sota and BC V proteins target interferon regulatory factor (IRF)-7 for degradation and promote the conversion of full-length IRF-7 to a lower molecular weight form (IRF-7*). This is the first demonstration that IRF-7 is targeted by a paramyxovirus V protein. The amount of IRF-7* decreases in a dose-dependent manner in the presence of a proteasome inhibitor, suggesting that IRF-7* is a degradation product of IRF-7. Furthermore, the BC V protein promotes complete conversion of IRF-7 to IRF7*, whereas the La Sota V protein does so less efficiently. Again, this is consistent with the difference in IFN antagonistic activity of the two V proteins, and in turn, with their virulence. The second aim is to characterize an HN-specific monoclonal antibody called AVS-I. A previous study suggested that AVS-I recognizes an epitope that is conserved in lentogenic strains and raises the possibility that this epitope may colocalize with a determinant of virulence in HN. To further characterize antibody AVS-I and the epitope it recognizes, we (i) determined its specificity for several additional strains of the virus, (ii) mapped its binding to HN in competition with our own antibodies, (iii) determined its functional inhibition profile, and (iv) isolated and sequenced an AVS-I escape mutant. The results demonstrate that AVS-I binds to a conformational epitope at the carboxy terminus of HN. This suggests that this region of HN may define a determinant of virulence. However, it was also shown that AVS-I, which was previously thought to be specific for avirulent strains of NDV, actually recognizes individual mesogenic and velogenic strains. In conclusion, the data presented in this dissertation contributes to a greater understanding of the molecular basis for NDV virulence and may aid in development of antiviral strategies and generation of recombinant NDVs suitable for use in cancer and gene therapy.
3

Characterization of the Relationship Between Measles Virus Fusion, Receptor Binding, and the Virus-Specific Interaction Between the Hemagglutinin and Fusion Glycoproteins: a Dissertation

Corey, Elizabeth Ann 17 May 2006 (has links)
Measles (MV) virions, like those of other enveloped viruses, enter cells by fusing their lipid membranes with those of the target host cells. Additionally, infected tissues often possess giant multinucleate cells, known as syncytia, which are formed by fusion of infected cells with uninfected neighbors. Expression of both the MV attachment (H) and fusion (F) proteins is required for membrane fusion. MV H mediates receptor binding in order to bring the two membranes into close proximity prior to F activation and is thought to trigger F activation through a specific interaction between the two proteins. Although measles H and F are efficiently transported to the cell surface when expressed independently, evidence has been reported in support of an intracellular interaction between the two proteins that can be detected using an ER co-retention approach. However, it was not determined if the putative co-retention was specific to the two measles glycoproteins, as is their ability to complement each other for efficient fusion promotion. Thus, in this thesis, the formation of an intracellular complex between MV H and F was re-examined. Consistent with the formation of an intracellular complex, cell surface expression and receptor binding of untagged wt MV H is slightly reduced by co-expression of an excess of ER-tagged MV F compared to co-expression with wt F. However, the reduction in surface expression is non-specific in that it can also be induced with heterologous proteins of NDV, which lack significant homology with those of MV. Although this approach did not detect a specific intracellular interaction between MV H and F, it cannot be ruled out that there is a weak association of the proteins that is undetectable by this method. This led to the use of an alternative approach to investigate the cellular site(s) of interaction between the measles H and F proteins. Consistent with a cell surface interaction between MV H and F, the combination of surface biotinylation and co-immunoprecipitation detects formation of a virus-specific H-F complex. Approximately, 21% of the total amount of MV H at the cell surface can be captured with MV F using an antibody against the latter protein. Two complementary approaches were used to address the relationship between this cell surface interaction and receptor recognition by MV H. First, the proteins were co-immunoprecipitated from the surface of Chinese hamster ovary (CHO) cells, which do not express either MV receptor, CD46 or CD150. Similar levels of MV H can be co-immunoprecipitated with F from the surfaces of parental CHO cells and stably transfected cells that express, human CD46 (CHO-CD46), indicating that binding to CD46 is not the trigger for the H-F interaction. Second, MV H proteins, carrying mutations that dramatically reduce CD46 binding, were shown to co-immunoprecipitate efficiently with F from the surface of HeLa cells. Significantly, these results indicate that MV H and F interact in the absence of, and thus prior to, receptor binding. This is in direct contrast to the NDV HN-F cell surface interaction, which is thought to be triggered by receptor binding. Identification of the domains of the para myxovirus attachment and fusion proteins that mediate membrane fusion activities is an essential part of understanding the mechanism of fusion. As a result of the H-F interaction prior to receptor binding, MV H attachment to its cellular receptor must result in conformational changes that trigger activation of the F protein. Site-directed mutagenesis analyses of two regions of MV H indicate that a HR domain in the stalk of the attachment protein is essential to the ability of H to activate F. However, either it is not the only region of H that interacts with F or it is indirectly involved in F activation because mutations in the HR do not disrupt MV H-F complex formation at the cell surface. Additionally, the functional interaction between MV H and F may be mediated, at least in part, by Loop 1 of the amino terminus of the C-rich region of the fusion protein. However, the exact role of this region of the F protein in fusion promotion remains to be determined. Importantly, the cell surface interaction between MV H and F proteins appears to be mediated by more that one region of each protein. In contrast to NDV, in no case has a definitive link between any single amino acid difference in MV H or F and an inability to form the cell surface H-F complex been established. In conclusion, the data presented in this dissertation support a model of measles membrane fusion in which the Hand F proteins form a complex prior to receptor recognition. This complex may hold F in its meta-stable pre-fusion state until binding of H to receptors at the cell surface triggers dissociation of the complex, releasing F to assume its fusogenic form. Importantly, these data also indicate that, although paramyxoviruses may all use the same general process. for promotion of membrane fusion, the mechanism may vary in multiple aspects. A more complete understanding of the means by which measles promotes membrane fusion may direct the development of specific strategies aimed at interfering with the early stages of infection.
4

Characterization of the Interaction Between the Attachment and Fusion Glycoproteins Required for Paramyxovirus Fusion: a Dissertation

Melanson, Vanessa R. 16 December 2005 (has links)
The first step of viral infection requires the binding of the viral attachment protein to cell surface receptors. Following binding, viruses penetrate the cellular membrane to deliver their genome into the host cell. For enveloped viruses, which have a lipid bilayer that surrounds their nucleocapsids, entry into the host cell requires the fusion of viral and cellular membranes. This process is mediated by viral glycoproteins located on the surface of the virus. For many enveloped viruses, such as influenza, Ebola, and human immunodeficiency virus, the fusion protein is responsible for mediating both attachment to cellular receptors and membrane fusion. However, paramyxoviruses are unique among fusion promoting viruses because their receptor binding and fusion activities reside on two separate proteins. This unique distribution of functions necessitates a mechanism by which the two proteins can transmit the juxtaposition of the viral and host cell membranes, mediated by the attachment protein (HN/H), into membrane fusion, mediated by the fusion (F) protein. This mechanism allows for paramyxoviruses to gain entry into and spread between cells, and therefore, is an important aspect of virus infection and disease progression. Despite the conservation of receptor binding activity among members of the Paramyxovirinaesubfamily, for most of these viruses, including Newcastle disease virus (NDV), heterologous HN proteins cannot complement F in the promotion of fusion; both the HN and F proteins must originate from the same virus. This is consistent with the existence of a virus-specific interaction between the two glycoproteins. Thus, one or more domains on the HN and F proteins is thought to mediate a specific interaction between them that is an integral part of the fusion process. Therefore, the primary focus of this thesis is the identification of the site(s) on HN that directly contacts F in the HN-F interaction. The ectodomain of the HN protein consists of a stalk and a terminal globular head. Analysis of the fusion activity of chimeric paramyxovirus HN proteins indicates that the stalk region of HN determines its F protein specificity. The first goal of this research was to address the question of whether the stalk not only determines F-specificity, but does so by directly mediating the interaction with F. To establish a correlation between the amount of fusion and the extent of the HN-F interaction, a specific and quantitative co-immunoprecipitation assay was used that detects the HN-F complex at the cell surface. As an initial probe of the role of the HN stalk in mediating the interaction with F, N-glycans were individually added at several positions in the region. N-glycan addition at positions 69 and 77 in the stalk specifically and completely block both fusion and the HN-F interaction without affecting either HN structure or its other activities. However, though they also prevent fusion, N-glycans added at other positions in the stalk also modulate activities that reside in the globular head of HN. This correlates with an alteration of the tetrameric structure of the protein as indicated by sucrose gradient sedimentation analyses. These additional N-glycans likely indirectly affect fusion, perhaps by interfering with changes in the conformation of HN that link receptor binding to the fusion activation of F. To address the issue of whether N-glycan addition at any position in HN would abolish fusion, an N-glycan was added in another region at the base of the globular head of HN (residues 124-152), which was previously predicted by a peptide-based analysis to mediate the interaction with F. HN carrying this additional N-glycan exhibits significant fusion promoting activity, arguing against this site being part of the F-interactive domain in HN. These data support the idea that the F-interactive site on HN is defined by the stalk region of the protein. Site-directed mutagenesis was used to begin to explore the role of individual residues in the stalk in the interaction with F. The characteristics of the F-interactive domain in the stalk of HN are that it is a conserved motif with enough sequence heterogeneity to account for the specificity of the interaction. One such region that meets these requirements is the intervening region (IR) (residues 89-95); a non-helical domain situated between two conserved heptad repeats. Several amino acid substitutions for a completely conserved proline residue in this region impair not only fusion and the HN-F interaction, but also decrease neuraminidase activity in the globular domain and alter the structure of the protein, suggesting that the substitutions indirectly affect the HN-F interaction. Substitutions for L94 also interfere with fusion, but have no significant effect on any other HN function or its structure. Amino acid substitutions at two other positions in the IR (A89 and L90) also modulate only fusion. In all cases, diminished fusion correlates with a decreased ability of the mutated HN protein to interact with F at the cell surface. These findings indicate that the IR is critical to the role of HN in the promotion of fusion and are consistent with its direct involvement in the interaction with the homologous F protein. These are the first point mutations in the HN protein for which a correlation has been demonstrated between the extent of the HN-F interaction and the amount of fusion. This argues strongly that the co-IP assay is an accurate reflection of the HN-F interaction. The second goal of this research was to address the HN-F interaction from the perspective of the F protein by investigating the relationship between receptor binding, the HN-F interaction, and fusion using a highly fusogenic form of the F protein. It has previously been shown that an L289A substitution in NDV F eliminates the requirement for HN in the promotion of fusion and enhances HN-dependent fusion above wild-type (wt) levels. Here, it was shown that the HN-independent fusion exhibited by L289A-F in Cos-7 cells cannot be duplicated in BHK cells. However, when L289A-F is co-expressed with wt HN, enhanced fusion above wt levels is observed in BHK cells. Additionally, when L289A-F is co-expressed with IR-mutated HN proteins previously shown to promote low levels of fusion with wt F, a 2.5-fold increase in fusion was observed. However, similar to wt F, an interaction between L289A-F and the IR-mutated HN proteins was not detected. These results imply that the attachment function of HN, as well as the conformational change in L289A-F, are necessary for the enhanced level of fusion exhibited by HN proteins co-expressed with L289A-F. Indeed, two MAbs detected a conformational difference between L289A-F and the wt F protein. These findings support the idea that the L289A substitution converts F to a form that is less dependent on an interaction with HN for conversion to the fusion-active form. The last goal of this research was to address the cellular site of the HN-F interaction, still a controversial issue based on conflicting data from studies of different paramyxoviruses, using various approaches. This is a particular point of interest, as it speaks to the mechanism by which the HN-F interaction regulates fusion. Thus, NDV HN and F were successfully retained intracellularly with a multiple arginine or KK motif, respectively. The results of Endoglycosidase H resistance and F cleavage studies indicate that the mutated proteins, HN-ER and F-ER, are retained in a compartment prior to the medial-Golgi apparatus and that they are unable to interact with a high enough affinity to co-retain or even cause reduced transport of their wt partner glycoproteins. This is consistent with the HN-F interaction occurring at the cell surface, possibly triggered by receptor binding. In conclusion, this thesis presents evidence to argue that the IR in the stalk of the NDV HN protein directly mediates the interaction with the F protein that is necessary for fusion. Overall, the data presented in this thesis extend the current knowledge of the mechanism by which the paramyxovirus attachment protein can trigger the F protein to initiate membrane fusion. A clear understanding of this process has the potential to identify new anti-viral strategies, such as small molecule inhibitors, aimed at controlling paramyxovirus infection by interfering with early steps in the virus infection cycle.
5

Role of Host Cellular Membrane Raft Domains in the Assembly and Release of Newcastle Disease Virus: A Dissertation

Laliberte, Jason P. 01 April 2008 (has links)
Newcastle disease virus (NDV) belongs to the Paramyxoviridae, a family of enveloped RNA viruses that includes many important human and animal pathogens. Although many aspects of the paramyxovirus life cycle are known in detail, our understanding of the mechanisms regulating paramyxovirus assembly and release are poorly understood. For many enveloped RNA viruses, it has recently become apparent that both viral and host cellular determinants coordinate the proper and efficient assembly of infectious progeny virions. Utilizing NDV as a model system to explore viral and cellular determinants of paramyxovirus assembly, we have shown that host cell membrane lipid raft domains serve as platforms of NDV assembly and release. This conclusion was supported by several key experimental results, including the exclusive incorporation of host cell membrane raftassociated molecules into virions, the association of structural components of the NDV particle with membrane lipid raft domains in infected cells and the strong correlation between the kinetics of viral protein dissociation from membrane lipid raft domains and incorporation into virions. Moreover, perturbation of infected cell membrane raft domains during virus assembly resulted in the disordered assembly of abnormal virions with reduced infectivity. These results further established membrane raft domains as sites of virus assembly and showed the integrity of these domains to be critical for the proper assembly of infectious virions. Although specific viral protein-protein interactions are thought to occur during paramyxovirus assembly, our understanding of how these interactions are coordinated is incomplete. While exploring the mechanisms underlying the disordered assembly of non-infectious virions in membrane raft-perturbed cells, we determined that the integrity of membrane raft domains was critical in the formation and virion incorporation of a complex consisting of the NDV attachment (HN) and fusion (F) proteins. The reduced virus-to-cell membrane fusion capacity of particles released from membrane raft-perturbed cells was attributed to an absence of the HN – F glycoprotein-containing complex within the virion envelope. This result also correlated with a reduction of these glycoprotein complexes in membrane lipid raft fractions of membrane raft-perturbed cells. Specifically, it was determined that the formation of newly synthesized HN and F polypeptides into the glycoprotein complex destined for virion incorporation was dependent on membrane lipid raft integrity. Finally, a novel virion complex between the ribonucleoprotein (RNP) structure and the HN attachment protein was identified and characterized. Unlike the glycoprotein complex, the detection of the RNP – HN protein-containing complex was not affected by membrane raft perturbation during virus assembly in the cell. The biological importance of this novel complex for the proper assembly of an infectious progeny virion is currently under investigation. The results presented in this thesis outline the role of host cell membrane lipid raft domains in the assembly and release processes of a model paramyxovirus. Furthermore, the present work extends our understanding of how these particular host cell domains mechanistically facilitate the ordered assembly and release of an enveloped RNA virus.
6

Untersuchungen zur F-proteinvermittelten Fusion von Paramyxoviren

Baljinnyam, Bolormaa 25 March 2003 (has links)
Die für die Vermehrung der Paramyxoviren notwendige Freisetzung des Virusgenoms in die Wirtszelle findet nach einer Verschmelzung der Virushülle mit der Zellmembran statt. Die Membranfusion wird durch eine Konformationsänderung des membranständigen Fusionsproteins (F-Protein) der Paramyxoviren vermittelt. Der Auslöser der Strukturumwandlung des F-Proteins ist bislang unbekannt. Man nimmt an, daß eine Wechselwirkung mit dem zweiten membranständigen Protein der Hämagglutinin-Neuraminidase (HN-Protein) die Strukturumwandlung des F-Proteins induziert. Das F-Protein kann jedoch auch in Abwesenheit des HN-Proteins eine Membranfusion vermitteln. Für das Verständnis des Mechanismus der F-proteinvermittelten Fusion ist die Kenntnis der dreidimensionalen Struktur des F-Proteins notwendig. In der vorliegenden Arbeit wurden die F-Proteine der Paramyxoviren, Sendaivirus und Simianvirus 5, in fusionskompetenter Form isoliert und in kleine Lipidvesikel rekonstituiert, um deren Struktur mittels Kryoelektronenmikroskopie und Einzelpartikelanalyse aufzuklären. Die 3D-Struktur des Sendaivirus-F-Proteins konnte mit einer Auflösung von 16 Angström aufgeklärt werden. Es ist die erste 3D-Struktur des F-Proteins eines Paramyxovirus in der fusionskompetenten Form. Um geeignete Bedingungen herauszufinden, die das Auslösen der Konformationsänderung der F-Proteine bzw. das "Einfangen" von Strukturintermediaten während der Fusion ermöglichen, wurde das Fusionsverhalten von Sendaivirus und Simianvirus 5 bei unterschiedlichen Temperatur- und pH-Werten sowie in Anwesenheit von Lysolipiden mittels Fluoreszenzdequenchingassays untersucht. Ein signifikanter Anstieg der Fusionsaktivität der untersuchten Viren konnte durch eine Erhöhung der Temperatur erreicht werden. Mittels ESR-Spektroskopie unter Einsatz von spinmarkierten Lysolipiden konnte gezeigt werden, daß Lysolipide die proteinvermittelte Fusion von Hüllviren in einem späten lipidabhängigen Schritt hemmen. Diese Untersuchungen bilden damit eine Grundlage zur Aufklärung der 3D-Struktur des F-Proteins im fusionsaktiven Zustand. Desweiteren wurde die Rolle der transmembranalen und zytoplasmatischen Domäne des F-Proteins bei der Membranfusion und der Wechselwirkung mit dem HN-Protein mittels Fluoreszenzmikroskopie untersucht. Die Befunde der 3D-Strukturaufklärung und der fluoreszenzmikroskopischen Studien wurden unter anderem in Hinblick auf die Bedeutung der Wechselwirkung zwischen den F- und HN-Proteinen für die Fusion diskutiert. / Paramyxoviruses infect their host cells by fusion of the viral envelope with the cell membrane. The membrane fusion is mediated by a confomational change of a viral envelope glycoprotein called the fusion (F) protein. The trigger of the F protein conformational change is still unknown. It is suggested, that an interaction of the F protein with the second envelope glycoprotein hemagglutinin-neuraminase (HN) induces its conformational change. However the F protein can mediate membrane fusion in absense of HN. The knowledge of the three dimensional structure of the F protein is reqiured to understand the F mediated membrane fusion. In the present work the fusion competent form of the fusion proteins of the paramyxoviruses Sendai virus and Simian virus 5 were isolated and incorporated each of them into small lipid vesicles. The 3D-structure of the entire ectodomain of the Sendai virus F protein has been determined in fusion potential conformation by cryo electron microscopy of single molecules and 3D-reconstruction at a resolution of 16 Angström. To detect usefull conditions for triggering the conformational change of F, the fusion of Sendai virus and Simian virus 5 have been studied at different temperature and pH, respectively, using a fluorescence dequenching assay. A significant increase of virus fusion activity has been found due to temperature enhancement. Using ESR-spectroscopy and spin-labeled lysolipids it has been shown that lysolipids inhibit the protein mediated fusion of enveloped viruses at a late lipid-dependent intermediate. Thus lysolipids are capable to freeze a conformational intermediate of the F protein during fusion. Furthermore the role of the transmembrane and the cytoplasmic domain of the Sendai virus F protein for membrane fusion was investigated using fluorescence microscopy. The results of the fluorescence microscopy study and the detection of the 3D-structure have been discussed in view of the relevance of F-HN-interaction for membrane fusion.

Page generated in 0.0662 seconds