The generation of native human monoclonal antibodies with neutralising activity for dengue virus 3

199 317 0
The generation of native human monoclonal antibodies with neutralising activity for dengue virus 3

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

Chapter - Introduction 1.1 Dengue virus 1.1.1 Classification Dengue viruses (DV) are members of the Flavivirus genus of the Flaviviridae family which comprises of over 70 members separated into groups using molecular phylogenetic analyses and analyses of serological relatedness. Viruses in the Flavivirus genus can cause serious diseases in humans and animals, and most of them are anthropod-borne (arboviruses) and thus are transmitted to vertebrate hosts by mosquitoes or ticks. Several members of the Flavivirus genus, such as Yellow Fever virus (YFV), West Nile virus (WNV), Japanese Encephalitis virus (JEV) and in particular Dengue virus (DV) are highly pathogenic to humans and constitute major international health problems (Mackenzie, Gubler et al. 2004; Gould and Solomon 2008). Within the genus, the viruses can be further subdivided into antigenic complexes according to serological criteria, or into clusters, clades and species on the basis of molecular phylogenetics (1.1.6) and is summarized in Table 1.1 (Kuno, Chang et al. 1998). Table.1.1. Flavivirus Classification. The above dendrogram shows the relationships between selected flaviviruses. (2001) Lippincott Williams and Wilkins, Philadelphia. In Fields Virology. Based on the gene sequence of a non-structural protein, NS5, the flaviviridae are clustered into three distinct groups which correlate with the mode of transmission (mosquito-borne, tick-borne, unknown vector respectively)(Kuno, Chang et al. 1998). DV was determined to be evolved from a common ancestor 1,000 years ago and that human transmission started between 125 and 325 years ago (Twiddy, Holmes et al. 2003; Mackenzie, Gubler et al. 2004). It is still undetermined whether the virus originated from Africa or from Asia but dengue transmission was first maintained in a sylvatic lifecycle within the Aedes species of mosquitoes (Twiddy, Holmes et al. 2003). Sporadic outbreaks of dengue fever first occurred in predomestic regions due to transmission by Aedes albopictus and Aedes aegypti. The adaptation of Aedes species, mainly the Aedes aegypti, to urban and densely inhabited areas is proposed to have created optimal conditions for human transmission resulting in the emergence of dengue epidemics (Gubler 2002; Gubler 2004; Mackenzie, Gubler et al. 2004). 1.1.2 Epidemiology At present, close to 2.5 billion people living in more than 100 dengue endemic countries in the tropical/sub-tropical belt are considered to be at risk of dengue infection (Pinheiro and Corber 1997). Approximately 50 million people are infected each year with DV with over 500 000 people requiring hospitalization. All four DV serotypes are infectious to humans. Severe disease was first detected in Southeast Asia and the Western Pacific region. Over the years, the geographic distribution has increased to include South Asia, South and Central America, the Caribbean, and Africa. This is mainly due to the increased accessibility to infected areas with the aid of modern transport. Over billion people live in endemic areas and therefore at risk of developing the disease, thus making dengue an emerging disease and global threat. The outbreaks in Hawaii, along the Texas-Mexico border and Puerto Rico make the widespread appearance of dengue a possibility in the United States (FIG.1.1). FIG.1.1. Global prevalence of DF and DHF as shown by WHO. World map showing the prevalence of DV in 2005. http://www.who.int/csr/disease/dengue/impact/en/index.html. All four DV serotypes are prevalent in Singapore. In 2006, DV1 remained the predominant serotype after the major 2004-2005 outbreak and more than 75 % of the samples are DV1 positive. In early January 2007, the predominant circulating serotype switched from DV1 to DV2. The proportion of DV2 positive samples rose from 57.9 % to 91 % at mid-2007. The rise of DV3 cases were detected in 2008. Early attempts to step up prevention of the spread of the serotype that had been uncommon in Singapore have been implemented to prevent another outbreak. The fatality rates are low in Singapore with 0.32 % in 2006 and 0.27 % in 2007 and rates remained unchanged the years after (Ooi, Goh et al. 2006). 1.1.3 Structure of dengue virions Members of the Flaviviridae family are characterized by having enveloped virions of small size containing three structural proteins, the envelope (E), core (C) and precursor membrane (prM) and seven non-structural (NS) proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5). The E, C and prM proteins have 495, 120 and 165 amino acids respectively (Mukhopadhyay, Kuhn et al. 2005). prM is processed to the mature M protein late in secretion in the trans Golgi compartment by furin (Stadler, Allison et al. 1997). Multiple copies of the C protein (11kDa) encapsulate the viral RNA genome to form the viral nucleocapsid (Chang, Luh et al. 2001; Jones, Ma et al. 2003; Ma, Jones et al. 2004). The E protein has three distinct structural domains termed domain I, II and III respectively (Rey, Heinz et al. 1995; Modis, Ogata et al. 2003; Modis, Ogata et al. 2005; Nybakken, Nelson et al. 2006). Domain I (DI) is structurally positioned between Domain II (DII), the homodimerizaton domain, and the immunoglobulinlike domain III (DIII) (FIG. 1.2). The mature dengue virion has a diameter of about 500Å and consist of a viral genome of around 10.8kb packed by the dimeric capsid proteins. The resulting nucleocapsid is enclosed by a host-derived lipid bilayer containing 180 copies of the E and M protein that form an icosahedral symmetry (T=3) (Kuhn, Zhang et al. 2002). This mean that the three E monomers present in each icosahedral asymmetric unit exist in three chemically distinct environments and may therefore play a distinct role in different stages of the infection. Based on the shape of the monomer and the location of the antibody epitopes, Rey et al. postulated that the E protein has a „flat‟ topology along the surface of the virus lipid bilayer (Rey, Heinz et al. 1995). FIG.1.2. Ribbon drawing of E protein. Dengue E protein dimer with three defined domains within each monomer: Domain I in red, Domain II in yellow with fusion loop in green, and Domain III in blue (Modis, Ogata et al. 2003). 1.1.4 Organization of the Flavivirus genome The viral genome consists of a single stranded, positive-sense RNA and approximately 10.8kb in length (Chambers, Hahn et al. 1990) (FIG.1.3). The genome has one open reading frame encoding a single polyprotein. The 5‟ end of the RNA contains a type I cap and is followed by the conserved dinucleotide sequence AG. The type I cap is generated when the first nucleotide in the transcript correspond to this position. Genomic RNA of mosquito-borne flaviviruses appears to lack a 3‟ poly-(A) tract and instead terminate with the conserved dinucleotide CU. The flaviviral genome contains an open reading frame of over 10000 bases, flanked by 5‟ and 3‟ untranslated regions (UTR) containing conserved RNA elements. No other conserved open reading frame (ORF) has been identified in either the genomic sense RNA or its compliment (Chambers, Hahn et al. 1990). The amino terminus of the genome encodes the prM, C and E proteins that constitute the virus particle. Seven NS proteins are essential for viral replication and are encoded by the remainder of the genome. The C protein is involved with packaging of the viral genome and forming the nucleocapsid (NC). prM and E are glycoproteins, each of which contains two transmembrane helices. Before it is cleaved during particle maturation to yield the pr peptide and the M protein (75 amino acids), the prM protein may function as a chaperone for folding of the E protein. The E protein contains a cellular receptor binding site and a fusion peptide (Stadler, Allison et al. 1997; Allison, Schalich et al. 2001; Lorenz, Allison et al. 2002). Since flaviviruses only encode 10 proteins, the host cell protein synthesis, nucleic acid synthesis, membrane trafficking machinery and functions are exploited in order to complete the viral infectious cycle. FIG.1.3. Schematic representation of the polyprotein processing for flaviviruses. The top region depicts the structural and non-structural ORF and the 5‟ and 3‟ UTR. The bottom region depicts co-translational cleavage by signalase and NS3 protease separating structural and non-structural proteins occurs at the C-terminus of E and the roles of virus proteins. Figure adapted from (FernandezGarcia, Mazzon et al. 2009) 1.1.5 Replication strategy 1.1.5.1 Receptor interaction Infection with one of the arthropod-borne flaviviruses begins when the vector takes a blood meal and the virus is introduced into the host. Despite a small number of reports suggesting other entry mechanisms of dengue virus such as entry via direct fusion with the plasma membrane (Hase, Summers et al. 1989; Lim and Ng 1999), the receptor-mediated endocytosis is generally accepted as the principle mode of entry. Autopsy studies have indicated the virus infects dendritic cells (DCs), monocytes/macrophages, B cells (Tassaneetrithep, Burgess et al. 2003), T cells, endothelial cells, hepatocytes and neuronal cells (Clyde, Kyle et al. 2006). Further evidence for this broad cellular tropism in vivo has included the detection of DV in Langerhans cells (skin-resident DCs) (Scott, Nisalak et al. 1980; King, Nisalak et al. 1999; Wu, Grouard-Vogel et al. 2000; Neves-Souza, Azeredo et al. 2005) after inoculation with an experimental dengue vaccine and in monocytes and B cells in peripheral blood from naturally infected patients (Scott, Nisalak et al. 1980; King, Nisalak et al. 1999; Wu, Grouard-Vogel et al. 2000; Neves-Souza, Azeredo et al. 2005). Several groups have attempted to characterize the cellular receptors of DV infection. A number of different mammalian cell receptors have been proposed. Evidence of heparin sulfate (Chen, Maguire et al. 1997), heat shock protein 70 (HSP70) and HSP90 (Reyes-Del Valle, ChavezSalinas et al. 2005), GRP78/BiP (Jindadamrongwech, Thepparit et al. 2004), CD14 (Chen, Wang et al. 1999), 37 kDa/67 kDa high affinity laminin receptor (Thepparit and Smith 2004), mannose receptor (MR) (Miller, de Wet et al. 2008), DC-specific intercellular adhesion molecule (ICAM-3)-grabbing nonintegrin (DC-SIGN, CD209) (Tassaneetrithep, Burgess et al. 2003) and liver/lymph nodespecific ICAM-3-grabbing nonintegrin (Tassaneetrithep, Burgess et al. 2003) have been provided. A growing body of evidence suggests that DC-SIGN provides a critical bridge between viral replication in insect vectors and infection of the vertebrate host though it should be noted that much of this datails based on human cell lines or animal models (Navarro-Sanchez, Altmeyer et al. 2003; Tassaneetrithep, Burgess et al. 2003; Lozach, Burleigh et al. 2005). DC-SIGN is a tetrameric C-type lectin that is unique for pathogen capture and antigen presentation (Cambi, Gijzen et al. 2003). This receptor is constitutively expressed on DCs, including Langerhans cells, the proposed cells present at the anatomical site of initial infection following the bite of a DV-infected mosquito (van Kooyk and Geijtenbeek 2003). The four DV serotypes of DV strains utilize DC-SIGN to enter into and infect productively immature DCs (Navarro-Sanchez, Altmeyer et al. 2003; Tassaneetrithep, Burgess et al. 2003; Martina, Koraka et al. 2008). The carbohydrate recognition domain (CRD) of DC-SIGN interacts with the N-glycosylated carbohydrate moieties of the E protein. DC-SIGN possesses the remarkable capacity to distinguish between high-mannose glycans typical of insect-derived glycoproteins and the complex glycosylation of host-derived proteins (van Kooyk and Geijtenbeek 2003; Lozach, Burleigh et al. 2005) implies that DV have evolved an elegant strategy to initiate infection of human cells by taking advantage of the ligand specificity of this pattern recognition receptor. A comparative study of two dengue strains showing differences in infectivity revealed an unique amino acid in the E-protein leading to the loss of N-linked glycosylation sites and therefore a decrease in infectivity (Ishak, Takegami et al. 2001). DC-SIGN-mediated infection may be an important component of DC maturation (Lozach, Burleigh et al. 2005), which is a crucial for allowing DCs to leave the skin and migrate to the lymph nodes, where they present processed antigens to T cells and initiate adaptive immune responses (Mellman and 10 Kaufman, B. M., P. L. Summers, et al. (1989). "Monoclonal antibodies for dengue virus prM glycoprotein protect mice against lethal dengue infection." Am J Trop Med Hyg 41(5): 576-580. Kaufmann, B., G. E. Nybakken, et al. (2006). "West Nile virus in complex with the Fab fragment of a neutralizing monoclonal antibody." Proc Natl Acad Sci U S A 103(33): 12400-12404. Kaye, K. M., K. M. Izumi, et al. (1993). "Epstein-Barr virus latent membrane protein is essential for B-lymphocyte growth transformation." Proc Natl Acad Sci U S A 90(19): 9150-9154. Kerr, B. M., A. L. Lear, et al. (1992). "Three transcriptionally distinct forms of EpsteinBarr virus latency in somatic cell hybrids: cell phenotype dependence of virus promoter usage." Virology 187(1): 189-201. Khanna, R., D. J. Moss, et al. (1999). "Vaccine strategies against Epstein-Barr virusassociated diseases: lessons from studies on cytotoxic T-cell-mediated immune regulation." Immunol Rev 170: 49-64. King, A. D., A. Nisalak, et al. (1999). "B cells are the principal circulating mononuclear cells infected by dengue virus." Southeast Asian J Trop Med Public Health 30(4): 718-728. Kinney, R. M. and C. Y. Huang (2001). "Development of new vaccines against dengue fever and Japanese encephalitis." Intervirology 44(2-3): 176-197. Klasse, P. J. and Q. J. Sattentau (2002). "Occupancy and mechanism in antibodymediated neutralization of animal viruses." J Gen Virol 83(Pt 9): 2091-2108. Klein, U. and R. Dalla-Favera (2008). "Germinal centres: role in B-cell physiology and malignancy." Nat Rev Immunol 8(1): 22-33. Klungthong, C., R. Putnak, et al. (2008). "Molecular genotyping of dengue viruses by phylogenetic analysis of the sequences of individual genes." J Virol Methods 154(1-2): 175-181. Kuhn, R. J., W. Zhang, et al. (2002). "Structure of dengue virus: implications for flavivirus organization, maturation, and fusion." Cell 108(5): 717-725. Kummerer, B. M. and C. M. Rice (2002). "Mutations in the yellow fever virus nonstructural protein NS2A selectively block production of infectious particles." J Virol 76(10): 4773-4784. Kuno, G., G. J. Chang, et al. (1998). "Phylogeny of the genus Flavivirus." J Virol 72(1): 7383. Kurosu, T., P. Chaichana, et al. (2007). "Secreted complement regulatory protein clusterin interacts with dengue virus nonstructural protein 1." Biochem Biophys Res Commun 362(4): 1051-1056. Kurth, J., M. L. Hansmann, et al. (2003). "Epstein-Barr virus-infected B cells expanding in germinal centers of infectious mononucleosis patients not participate in the germinal center reaction." Proc Natl Acad Sci U S A 100(8): 4730-4735. Kurth, J., T. Spieker, et al. (2000). "EBV-infected B cells in infectious mononucleosis: viral strategies for spreading in the B cell compartment and establishing latency." Immunity 13(4): 485-495. LaFleur, C., J. Granados, et al. (2002). "HLA-DR antigen frequencies in Mexican patients with dengue virus infection: HLA-DR4 as a possible genetic resistance factor for dengue hemorrhagic fever." Hum Immunol 63(11): 1039-1044. Lai, C. J., A. P. Goncalvez, et al. (2007). "Epitope determinants of a chimpanzee dengue virus type (DENV-4)-neutralizing antibody and protection against DENV-4 184 challenge in mice and rhesus monkeys by passively transferred humanized antibody." J Virol 81(23): 12766-12774. Laichalk, L. L., D. Hochberg, et al. (2002). "The dispersal of mucosal memory B cells: evidence from persistent EBV infection." Immunity 16(5): 745-754. Lanier, L. L. (2008). "Up on the tightrope: natural killer cell activation and inhibition." Nat Immunol 9(5): 495-502. Leng, C. H., S. J. Liu, et al. (2009). "A novel dengue vaccine candidate that induces crossneutralizing antibodies and memory immunity." Microbes Infect 11(2): 288-295. Lescar, J., A. Roussel, et al. (2001). "The Fusion glycoprotein shell of Semliki Forest virus: an icosahedral assembly primed for fusogenic activation at endosomal pH." Cell 105(1): 137-148. Li, H., S. Clum, et al. (1999). "The serine protease and RNA-stimulated nucleoside triphosphatase and RNA helicase functional domains of dengue virus type NS3 converge within a region of 20 amino acids." J Virol 73(4): 3108-3116. Libraty, D. H., T. P. Endy, et al. (2002). "Differing influences of virus burden and immune activation on disease severity in secondary dengue-3 virus infections." J Infect Dis 185(9): 1213-1221. Lim, H. Y. and M. L. Ng (1999). "A different mode of entry by dengue-2 neutralisation escape mutant virus." Arch Virol 144(5): 989-995. Lin, C. F., S. C. Chiu, et al. (2005). "Expression of cytokine, chemokine, and adhesion molecules during endothelial cell activation induced by antibodies against dengue virus nonstructural protein 1." J Immunol 174(1): 395-403. Lin, C. F., H. Y. Lei, et al. (2003). "Antibodies from dengue patient sera cross-react with endothelial cells and induce damage." J Med Virol 69(1): 82-90. Lin, S. R., S. C. Hsieh, et al. (2004). "Study of sequence variation of dengue type virus in naturally infected mosquitoes and human hosts: implications for transmission and evolution." J Virol 78(22): 12717-12721. Lindenbach, B. D. and C. M. Rice (2003). "Molecular biology of flaviviruses." Adv Virus Res 59: 23-61. Liu, W. J., H. B. Chen, et al. (2003). "Molecular and functional analyses of Kunjin virus infectious cDNA clones demonstrate the essential roles for NS2A in virus assembly and for a nonconservative residue in NS3 in RNA replication." J Virol 77(14): 7804-7813. Liu, Y. J. and C. Arpin (1997). "Germinal center development." Immunol Rev 156: 111126. Lok, S. M., V. Kostyuchenko, et al. (2008). "Binding of a neutralizing antibody to dengue virus alters the arrangement of surface glycoproteins." Nat Struct Mol Biol 15(3): 312-317. Lok, S. M., M. L. Ng, et al. (2001). "Amino acid and phenotypic changes in dengue virus associated with escape from neutralisation by IgM antibody." J Med Virol 65(2): 315-323. Loke, H., D. Bethell, et al. (2002). "Susceptibility to dengue hemorrhagic fever in vietnam: evidence of an association with variation in the vitamin d receptor and Fc gamma receptor IIa genes." Am J Trop Med Hyg 67(1): 102-106. Loke, H., D. B. Bethell, et al. (2001). "Strong HLA class I--restricted T cell responses in dengue hemorrhagic fever: a double-edged sword?" J Infect Dis 184(11): 13691373. 185 Longnecker, R. (2000). "Epstein-Barr virus latency: LMP2, a regulator or means for Epstein-Barr virus persistence?" Adv Cancer Res 79: 175-200. Lorenz, I. C., S. L. Allison, et al. (2002). "Folding and dimerization of tick-borne encephalitis virus envelope proteins prM and E in the endoplasmic reticulum." J Virol 76(11): 5480-5491. Lozach, P. Y., L. Burleigh, et al. (2005). "Dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN)-mediated enhancement of dengue virus infection is independent of DC-SIGN internalization signals." J Biol Chem 280(25): 23698-23708. Ma, L., C. T. Jones, et al. (2004). "Solution structure of dengue virus capsid protein reveals another fold." Proc Natl Acad Sci U S A 101(10): 3414-3419. Mackenzie, J. (2005). "Wrapping things up about virus RNA replication." Traffic 6(11): 967-977. Mackenzie, J. M., A. A. Khromykh, et al. (1998). "Subcellular localization and some biochemical properties of the flavivirus Kunjin nonstructural proteins NS2A and NS4A." Virology 245(2): 203-215. Mackenzie, J. S., D. J. Gubler, et al. (2004). "Emerging flaviviruses: the spread and resurgence of Japanese encephalitis, West Nile and dengue viruses." Nat Med 10(12 Suppl): S98-109. MacLennan, I. C. (1994). "Germinal centers." Annu Rev Immunol 12: 117-139. Mangada, M. M. and A. L. Rothman (2005). "Altered cytokine responses of denguespecific CD4+ T cells to heterologous serotypes." J Immunol 175(4): 2676-2683. Martina, B. E., P. Koraka, et al. (2008). "DC-SIGN enhances infection of cells with glycosylated West Nile virus in vitro and virus replication in human dendritic cells induces production of IFN-alpha and TNF-alpha." Virus Res 135(1): 64-71. Mathew, A. and A. L. Rothman (2008). "Understanding the contribution of cellular immunity to dengue disease pathogenesis." Immunol Rev 225: 300-313. Matusan, A. E., M. J. Pryor, et al. (2001). "Mutagenesis of the Dengue virus type NS3 protein within and outside helicase motifs: effects on enzyme activity and virus replication." J Virol 75(20): 9633-9643. McArthur, J. H., A. P. Durbin, et al. (2008). "Phase I clinical evaluation of rDEN4Delta30200,201: a live attenuated dengue vaccine candidate designed for decreased hepatotoxicity." Am J Trop Med Hyg 79(5): 678-684. McBride, W. J. and H. Bielefeldt-Ohmann (2000). "Dengue viral infections; pathogenesis and epidemiology." Microbes Infect 2(9): 1041-1050. Meijer, C. J., N. M. Jiwa, et al. (1996). "Epstein-Barr virus and human T-cell lymphomas." Semin Cancer Biol 7(4): 191-196. Mellman, I. and R. M. Steinman (2001). "Dendritic cells: specialized and regulated antigen processing machines." Cell 106(3): 255-258. Messer, W. B., D. J. Gubler, et al. (2003). "Emergence and global spread of a dengue serotype 3, subtype III virus." Emerg Infect Dis 9(7): 800-809. Miller, J. L., B. J. de Wet, et al. (2008). "The mannose receptor mediates dengue virus infection of macrophages." PLoS Pathog 4(2): e17. Miller, S., S. Kastner, et al. (2007). "The non-structural protein 4A of dengue virus is an integral membrane protein inducing membrane alterations in a 2K-regulated manner." J Biol Chem 282(12): 8873-8882. 186 Mitchell, D. A., A. J. Fadden, et al. (2001). "A novel mechanism of carbohydrate recognition by the C-type lectins DC-SIGN and DC-SIGNR. Subunit organization and binding to multivalent ligands." J Biol Chem 276(31): 28939-28945. Modis, Y., S. Ogata, et al. (2003). "A ligand-binding pocket in the dengue virus envelope glycoprotein." Proc Natl Acad Sci U S A 100(12): 6986-6991. Modis, Y., S. Ogata, et al. (2004). "Structure of the dengue virus envelope protein after membrane fusion." Nature 427(6972): 313-319. Modis, Y., S. Ogata, et al. (2005). "Variable surface epitopes in the crystal structure of dengue virus type envelope glycoprotein." J Virol 79(2): 1223-1231. Mongkolsapaya, J., W. Dejnirattisai, et al. (2003). "Original antigenic sin and apoptosis in the pathogenesis of dengue hemorrhagic fever." Nat Med 9(7): 921-927. Mongkolsapaya, J., T. Duangchinda, et al. (2006). "T cell responses in dengue hemorrhagic fever: are cross-reactive T cells suboptimal?" J Immunol 176(6): 3821-3829. Morens, D. M. (1994). "Antibody-dependent enhancement of infection and the pathogenesis of viral disease." Clin Infect Dis 19(3): 500-512. Morens, D. M. and S. B. Halstead (1990). "Measurement of antibody-dependent infection enhancement of four dengue virus serotypes by monoclonal and polyclonal antibodies." J Gen Virol 71 ( Pt 12): 2909-2914. Mota, J., J. Ramos-Castaneda, et al. (2002). "Phylogenetic analysis of the envelope protein (domain III) of dengue viruses." Salud Publica Mex 44(3): 228-236. Mukhopadhyay, S., B. S. Kim, et al. (2003). "Structure of West Nile virus." Science 302(5643): 248. Mukhopadhyay, S., R. J. Kuhn, et al. (2005). "A structural perspective of the flavivirus life cycle." Nat Rev Microbiol 3(1): 13-22. Munoz-Jordan, J. L., G. G. Sanchez-Burgos, et al. (2003). "Inhibition of interferon signaling by dengue virus." Proc Natl Acad Sci U S A 100(24): 14333-14338. Muramatsu, M., K. Kinoshita, et al. (2000). "Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme." Cell 102(5): 553-563. Myat Thu, H., K. Lowry, et al. (2005). "Lineage extinction and replacement in dengue type virus populations are due to stochastic events rather than to natural selection." Virology 336(2): 163-172. Navarro-Sanchez, E., R. Altmeyer, et al. (2003). "Dendritic-cell-specific ICAM3-grabbing non-integrin is essential for the productive infection of human dendritic cells by mosquito-cell-derived dengue viruses." EMBO Rep 4(7): 723-728. Neuberger, M. S. (2008). "Antibody diversification by somatic mutation: from Burnet onwards." Immunol Cell Biol 86(2): 124-132. Neuhierl, B., R. Feederle, et al. (2002). "Glycoprotein gp110 of Epstein-Barr virus determines viral tropism and efficiency of infection." Proc Natl Acad Sci U S A 99(23): 15036-15041. Neves-Souza, P. C., E. L. Azeredo, et al. (2005). "Inducible nitric oxide synthase (iNOS) expression in monocytes during acute Dengue Fever in patients and during in vitro infection." BMC Infect Dis 5: 64. Niedobitek, G., A. Agathanggelou, et al. (1995). "Heterogeneous expression of EpsteinBarr virus latent proteins in endemic Burkitt's lymphoma." Blood 86(2): 659-665. Nimmerjahn, F. and J. V. Ravetch (2005). "Divergent immunoglobulin g subclass activity through selective Fc receptor binding." Science 310(5753): 1510-1512. 187 Nitsche, F., A. Bell, et al. (1997). "Epstein-Barr virus leader protein enhances EBNA-2mediated transactivation of latent membrane protein expression: a role for the W1W2 repeat domain." J Virol 71(9): 6619-6628. Nybakken, G. E., C. A. Nelson, et al. (2006). "Crystal structure of the West Nile virus envelope glycoprotein." J Virol 80(23): 11467-11474. Nybakken, G. E., T. Oliphant, et al. (2005). "Structural basis of West Nile virus neutralization by a therapeutic antibody." Nature 437(7059): 764-769. Ocampo, C. B. and D. M. Wesson (2004). "Population dynamics of Aedes aegypti from a dengue hyperendemic urban setting in Colombia." Am J Trop Med Hyg 71(4): 506-513. Okazaki, I. M., H. Hiai, et al. (2003). "Constitutive expression of AID leads to tumorigenesis." J Exp Med 197(9): 1173-1181. Oliphant, T., G. E. Nybakken, et al. (2006). "Antibody recognition and neutralization determinants on domains I and II of West Nile Virus envelope protein." J Virol 80(24): 12149-12159. Ooi, E. E., K. T. Goh, et al. (2006). "Dengue prevention and 35 years of vector control in Singapore." Emerg Infect Dis 12(6): 887-893. Pacsa, A. S., R. Agarwal, et al. (2000). "Role of interleukin-12 in patients with dengue hemorrhagic fever." FEMS Immunol Med Microbiol 28(2): 151-155. Pasqualucci, L., P. Neumeister, et al. (2001). "Hypermutation of multiple protooncogenes in B-cell diffuse large-cell lymphomas." Nature 412(6844): 341-346. Pearson, G., F. Dewey, et al. (1970). "Relation between neutralization of Epstein-Barr virus and antibodies to cell-membrane antigens-induced by the virus." J Natl Cancer Inst 45(5): 989-995. Perez, A. B., G. Garcia, et al. (2004). "IL-10 levels in Dengue patients: some findings from the exceptional epidemiological conditions in Cuba." J Med Virol 73(2): 230-234. Pham, P., R. Bransteitter, et al. (2003). "Processive AID-catalysed cytosine deamination on single-stranded DNA simulates somatic hypermutation." Nature 424(6944): 103-107. Phillips, J. T., G. Rice, et al. (2004). "A multicenter, open-label, phase II study of the immunogenicity and safety of a new prefilled syringe (liquid) formulation of Avonex in patients with multiple sclerosis." Clin Ther 26(4): 511-521. Pierson, T. C., Q. Xu, et al. (2007). "The stoichiometry of antibody-mediated neutralization and enhancement of West Nile virus infection." Cell Host Microbe 1(2): 135-145. Pinheiro, F. P. and S. J. Corber (1997). "Global situation of dengue and dengue haemorrhagic fever, and its emergence in the Americas." World Health Stat Q 50(3-4): 161-169. Pokidysheva, E., Y. Zhang, et al. (2006). "Cryo-EM reconstruction of dengue virus in complex with the carbohydrate recognition domain of DC-SIGN." Cell 124(3): 485-493. Polizel, J. R., D. Bueno, et al. (2004). "Association of human leukocyte antigen DQ1 and dengue fever in a white Southern Brazilian population." Mem Inst Oswaldo Cruz 99(6): 559-562. Pryor, M. J., J. M. Carr, et al. (2001). "Replication of dengue virus type in human monocyte-derived macrophages: comparisons of isolates and recombinant viruses with substitutions at amino acid 390 in the envelope glycoprotein." Am J Trop Med Hyg 65(5): 427-434. 188 Ramiro, A. R., P. Stavropoulos, et al. (2003). "Transcription enhances AID-mediated cytidine deamination by exposing single-stranded DNA on the nontemplate strand." Nat Immunol 4(5): 452-456. Ray, D. and P. Y. Shi (2006). "Recent advances in flavivirus antiviral drug discovery and vaccine development." Recent Patents Anti-Infect Drug Disc 1(1): 45-55. Revy, P., T. Muto, et al. (2000). "Activation-induced cytidine deaminase (AID) deficiency causes the autosomal recessive form of the Hyper-IgM syndrome (HIGM2)." Cell 102(5): 565-575. Rey, F. A., F. X. Heinz, et al. (1995). "The envelope glycoprotein from tick-borne encephalitis virus at A resolution." Nature 375(6529): 291-298. Reyes-Del Valle, J., S. Chavez-Salinas, et al. (2005). "Heat shock protein 90 and heat shock protein 70 are components of dengue virus receptor complex in human cells." J Virol 79(8): 4557-4567. Rico-Hesse, R. (2003). "Microevolution and virulence of dengue viruses." Adv Virus Res 59: 315-341. Robertson, E. S., T. Ooka, et al. (1996). "Epstein-Barr virus vectors for gene delivery to B lymphocytes." Proc Natl Acad Sci U S A 93(21): 11334-11340. Rogozin, I. B., L. M. Iyer, et al. (2007). "Evolution and diversification of lamprey antigen receptors: evidence for involvement of an AID-APOBEC family cytosine deaminase." Nat Immunol 8(6): 647-656. Roosendaal, J., E. G. Westaway, et al. (2006). "Regulated cleavages at the West Nile virus NS4A-2K-NS4B junctions play a major role in rearranging cytoplasmic membranes and Golgi trafficking of the NS4A protein." J Virol 80(9): 4623-4632. Rosen, L. (1986). "Dengue in Greece in 1927 and 1928 and the pathogenesis of dengue hemorrhagic fever: new data and a different conclusion." Am J Trop Med Hyg 35(3): 642-653. Rowe, M., D. T. Rowe, et al. (1987). "Differences in B cell growth phenotype reflect novel patterns of Epstein-Barr virus latent gene expression in Burkitt's lymphoma cells." EMBO J 6(9): 2743-2751. Sakuntabhai, A., C. Turbpaiboon, et al. (2005). "A variant in the CD209 promoter is associated with severity of dengue disease." Nat Genet 37(5): 507-513. Sarma, V., Z. Lin, et al. (1995). "Activation of the B-cell surface receptor CD40 induces A20, a novel zinc finger protein that inhibits apoptosis." J Biol Chem 270(21): 12343-12346. Schieffelin, J. S., J. M. Costin, et al. (2010). "Neutralizing and non-neutralizing monoclonal antibodies against dengue virus E protein derived from a naturally infected patient." Virol J 7: 28. Schlesinger, J. J., M. W. Brandriss, et al. (1987). "Protection of mice against dengue virus encephalitis by immunization with the dengue virus non-structural glycoprotein NS1." J Gen Virol 68 ( Pt 3): 853-857. Schul, W., W. Liu, et al. (2007). "A dengue fever viremia model in mice shows reduction in viral replication and suppression of the inflammatory response after treatment with antiviral drugs." J Infect Dis 195(5): 665-674. Scott, R. M., K. H. Eckels, et al. (1983). "Dengue vaccine: dose response in volunteers in relation to yellow fever immune status." J Infect Dis 148(6): 1055-1060. Scott, R. M., S. Nimmannitya, et al. (1976). "Shock syndrome in primary dengue infections." Am J Trop Med Hyg 25(6): 866-874. 189 Scott, R. M., A. Nisalak, et al. (1980). "Isolation of dengue viruses from peripheral blood leukocytes of patients with hemorrhagic fever." J Infect Dis 141(1): 1-6. Seah, C. L., V. T. Chow, et al. (1995). "Semi-nested PCR using NS3 primers for the detection and typing of dengue viruses in clinical serum specimens." Clin Diagn Virol 4(2): 113-120. Seah, C. L., V. T. Chow, et al. (1995). "Rapid, single-step RT-PCR typing of dengue viruses using five NS3 gene primers." J Virol Methods 51(2-3): 193-200. Shannon-Lowe, C., G. Baldwin, et al. (2005). "Epstein-Barr virus-induced B-cell transformation: quantitating events from virus binding to cell outgrowth." J Gen Virol 86(Pt 11): 3009-3019. Shresta, S., J. L. Kyle, et al. (2004). "Early activation of natural killer and B cells in response to primary dengue virus infection in A/J mice." Virology 319(2): 262273. Shresta, S., K. L. Sharar, et al. (2006). "Murine model for dengue virus-induced lethal disease with increased vascular permeability." J Virol 80(20): 10208-10217. Shresta, S., K. L. Sharar, et al. (2005). "Critical roles for both STAT1-dependent and STAT1-independent pathways in the control of primary dengue virus infection in mice." J Immunol 175(6): 3946-3954. Shrestha, B., J. D. Brien, et al. (2010). "The development of therapeutic antibodies that neutralize homologous and heterologous genotypes of dengue virus type 1." PLoS Pathog 6(4): e1000823. Simmons, C. P., T. Dong, et al. (2005). "Early T-cell responses to dengue virus epitopes in Vietnamese adults with secondary dengue virus infections." J Virol 79(9): 56655675. Sinclair, A. J., I. Palmero, et al. (1994). "EBNA-2 and EBNA-LP cooperate to cause G0 to G1 transition during immortalization of resting human B lymphocytes by Epstein-Barr virus." EMBO J 13(14): 3321-3328. Sohail, A., J. Klapacz, et al. (2003). "Human activation-induced cytidine deaminase causes transcription-dependent, strand-biased C to U deaminations." Nucleic Acids Res 31(12): 2990-2994. Stadler, K., S. L. Allison, et al. (1997). "Proteolytic activation of tick-borne encephalitis virus by furin." J Virol 71(11): 8475-8481. Stephens, H. A., R. Klaythong, et al. (2002). "HLA-A and -B allele associations with secondary dengue virus infections correlate with disease severity and the infecting viral serotype in ethnic Thais." Tissue Antigens 60(4): 309-318. Stiasny, K., S. Brandler, et al. (2007). "Probing the flavivirus membrane fusion mechanism by using monoclonal antibodies." J Virol 81(20): 11526-11531. Stiasny, K., S. Kiermayr, et al. (2006). "Cryptic properties of a cluster of dominant flavivirus cross-reactive antigenic sites." J Virol 80(19): 9557-9568. Sukupolvi-Petty, S., S. K. Austin, et al. (2007). "Type- and subcomplex-specific neutralizing antibodies against domain III of dengue virus type envelope protein recognize adjacent epitopes." J Virol 81(23): 12816-12826. Talavera, D., A. M. Castillo, et al. (2004). "IL8 release, tight junction and cytoskeleton dynamic reorganization conducive to permeability increase are induced by dengue virus infection of microvascular endothelial monolayers." J Gen Virol 85(Pt 7): 1801-1813. Tan, G. K. and S. Alonso (2009). "Pathogenesis and prevention of dengue virus infection: state-of-the-art." Curr Opin Infect Dis 22(3): 302-308. 190 Tan, G. K., J. K. Ng, et al. (2010). "A non mouse-adapted dengue virus strain as a new model of severe dengue infection in AG129 mice." PLoS Negl Trop Dis 4(4): e672. Tassaneetrithep, B., T. H. Burgess, et al. (2003). "DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells." J Exp Med 197(7): 823-829. Thepparit, C. and D. R. Smith (2004). "Serotype-specific entry of dengue virus into liver cells: identification of the 37-kilodalton/67-kilodalton high-affinity laminin receptor as a dengue virus serotype receptor." J Virol 78(22): 12647-12656. Thisyakorn, U. and S. Nimmannitya (1993). "Nutritional status of children with dengue hemorrhagic fever." Clin Infect Dis 16(2): 295-297. Tobollik, S., L. Meyer, et al. (2006). "Epstein-Barr virus nuclear antigen inhibits AID expression during EBV-driven B-cell growth." Blood 108(12): 3859-3864. Traggiai, E., S. Becker, et al. (2004). "An efficient method to make human monoclonal antibodies from memory B cells: potent neutralization of SARS coronavirus." Nat Med 10(8): 871-875. Twiddy, S. S., E. C. Holmes, et al. (2003). "Inferring the rate and time-scale of dengue virus evolution." Mol Biol Evol 20(1): 122-129. Twiddy, S. S., C. H. Woelk, et al. (2002). "Phylogenetic evidence for adaptive evolution of dengue viruses in nature." J Gen Virol 83(Pt 7): 1679-1689. Umareddy, I., K. F. Tang, et al. (2008). "Dengue virus regulates type I interferon signalling in a strain-dependent manner in human cell lines." J Gen Virol 89(Pt 12): 3052-3062. van der Most, R. G., J. Corver, et al. (1999). "Mutagenesis of the RGD motif in the yellow fever virus 17D envelope protein." Virology 265(1): 83-95. van der Schaar, H. M., M. J. Rust, et al. (2008). "Dissecting the cell entry pathway of dengue virus by single-particle tracking in living cells." PLoS Pathog 4(12): e1000244. van Kooyk, Y. and T. B. Geijtenbeek (2003). "DC-SIGN: escape mechanism for pathogens." Nat Rev Immunol 3(9): 697-709. Vaughn, D. W. (2000). "Invited commentary: Dengue lessons from Cuba." Am J Epidemiol 152(9): 800-803. Vaughn, D. W., S. Green, et al. (2000). "Dengue viremia titer, antibody response pattern, and virus serotype correlate with disease severity." J Infect Dis 181(1): 2-9. Wang, C. L., R. A. Harper, et al. (2004). "Genome-wide somatic hypermutation." Proc Natl Acad Sci U S A 101(19): 7352-7356. Wang, F., C. Gregory, et al. (1990). "Epstein-Barr virus latent membrane protein (LMP1) and nuclear proteins and 3C are effectors of phenotypic changes in B lymphocytes: EBNA-2 and LMP1 cooperatively induce CD23." J Virol 64(5): 23092318. Wang, F., C. D. Gregory, et al. (1987). "Epstein-Barr virus nuclear antigen specifically induces expression of the B-cell activation antigen CD23." Proc Natl Acad Sci U S A 84(10): 3452-3456. Wang, F., S. F. Tsang, et al. (1990). "Epstein-Barr virus nuclear antigen transactivates latent membrane protein LMP1." J Virol 64(7): 3407-3416. Wang, R., J. Epstein, et al. (2001). "Induction of CD4(+) T cell-dependent CD8(+) type responses in humans by a malaria DNA vaccine." Proc Natl Acad Sci U S A 98(19): 10817-10822. 191 Wang, W. K., S. R. Lin, et al. (2002). "Dengue type virus in plasma is a population of closely related genomes: quasispecies." J Virol 76(9): 4662-4665. Wang, W. K., T. L. Sung, et al. (2002). "Sequence diversity of the capsid gene and the nonstructural gene NS2B of dengue-3 virus in vivo." Virology 303(1): 181-191. Warke, R. V., K. Xhaja, et al. (2003). "Dengue virus induces novel changes in gene expression of human umbilical vein endothelial cells." J Virol 77(21): 1182211832. Weiss, L. M., L. A. Movahed, et al. (1989). "Detection of Epstein-Barr viral genomes in Reed-Sternberg cells of Hodgkin's disease." N Engl J Med 320(8): 502-506. Welsch, S., S. Miller, et al. (2009). "Composition and three-dimensional architecture of the dengue virus replication and assembly sites." Cell Host Microbe 5(4): 365375. Wengler, G., G. Czaya, et al. (1991). "In vitro synthesis of West Nile virus proteins indicates that the amino-terminal segment of the NS3 protein contains the active centre of the protease which cleaves the viral polyprotein after multiple basic amino acids." J Gen Virol 72 ( Pt 4): 851-858. Wilson, J. B., J. L. Bell, et al. (1996). "Expression of Epstein-Barr virus nuclear antigen-1 induces B cell neoplasia in transgenic mice." EMBO J 15(12): 3117-3126. Wu, S. J., G. Grouard-Vogel, et al. (2000). "Human skin Langerhans cells are targets of dengue virus infection." Nat Med 6(7): 816-820. Young, L. S., C. W. Dawson, et al. (2000). "The expression and function of Epstein-Barr virus encoded latent genes." Mol Pathol 53(5): 238-247. Young, L. S. and A. B. Rickinson (2004). "Epstein-Barr virus: 40 years on." Nat Rev Cancer 4(10): 757-768. Yu, I. M., W. Zhang, et al. (2008). "Structure of the immature dengue virus at low pH primes proteolytic maturation." Science 319(5871): 1834-1837. Yuan, J., E. Cahir-McFarland, et al. (2006). "Virus and cell RNAs expressed during Epstein-Barr virus replication." J Virol 80(5): 2548-2565. Zellweger, R. M., T. R. Prestwood, et al. (2010). "Enhanced infection of liver sinusoidal endothelial cells in a mouse model of antibody-induced severe dengue disease." Cell Host Microbe 7(2): 128-139. Zhang, C., M. P. Mammen, Jr., et al. (2005). "Clade replacements in dengue virus serotypes and are associated with changing serotype prevalence." J Virol 79(24): 15123-15130. Zhang, W., P. R. Chipman, et al. (2003). "Visualization of membrane protein domains by cryo-electron microscopy of dengue virus." Nat Struct Biol 10(11): 907-912. Zhang, Y., J. Corver, et al. (2003). "Structures of immature flavivirus particles." EMBO J 22(11): 2604-2613. Zivna, I., S. Green, et al. (2002). "T cell responses to an HLA-B*07-restricted epitope on the dengue NS3 protein correlate with disease severity." J Immunol 168(11): 5959-5965. 192 Chapter 10 - Appendix 2.1. Reaction mix for serotyping of dengue patients. Component Volume per reaction (ul) 1ug Total RNA 20 2uM gene specific primer (GSP) 10mM DNTP Mix DEPC-Treated Water 10 Enzyme 5X Reaction Mix 10 RNase Inhibitor Final Volume 50 2.4 Reaction mix for PCR of antibody heavy and light chains. Component Volume per reaction (ul) 1ug Total RNA 2uM gene specific primer (GSP) 10mM DNTP Mix DEPC-Treated Water Final Volume 10 Component Volume per reaction (ul) 10X RT Buffer 25mM MgCl2 0.1M DTT RNase Out (40U/ml) SuperScript III RT (200U/ul) Final Volume 10 193 2.5 Reaction mix for cDNA synthesis of Human Immunoglobulin Genes Component Volume per reaction (ul) 10XPCR Mix 50mM MgCl2 1.5 10mM DNTP Mix 10uM sense primer 10uM antisense primer Taq Pol (5U/ul) 0.4 cDNA Water 38.1 Final Volume 50.0 2.6 Reaction mix for attachment of linker to heavy and light chains Component Volume per reaction (ul) -ve 10XPCR Mix 50mM MgCl2 1.5 1.5 10mM DNTP Mix 10uM sense primer 10uM antisense primer 0.4 0.4 100ng DNA H chain - 100ng DNA L chain - Water 38.1 40.1 Final Volume 50.0 50.0 Taq Pol (5U/ul) 194 2.18.2 Reaction Mix for Scfv digestion Reaction mix for Not digestion was as follows: Vol (ul) Not I (10 000U/ml) NEB 2.0 10X BSA 10.0 10X Buffer 10.0 DNA (1µg) H2O Top up to 100ul Total Volume 100.0ul Reaction mix for Sfi1 digestion was as follows: Vol (ul) Sfi I (20 000U/ml) NEB 1.0 10X BSA 10.0 10X Buffer 10.0 DNA 50.0 H2O Top up to 100ul Total Volume 100.0ul Reaction mix for ligation was as follows: T4 DNA Ligase (400 000U/ml) NEB 10X T4 DNA Ligase Buffer DNA (insert) pCANTAB 5E (vector) H2O Total Volume Sample 1.0 -ve 1.0 DO1 (+ve) 1.0 2.0 20ng 150ng Top up to 20ul 20.0ul 2.0 150ng Top up to 20ul 20.0ul 2.0 20ng 150ng Top up to 20ul 20.0ul 195 2.19.1 Reaction mix for heavy and light chain amplification Reaction components 1x reaction mix 4x Master mix 5x iProof HF buffer 10μL 40μL 10mM dNTP mix μL 4μL Upstream Primer [10μM] 2.5μL 10μL Downstream Primer [10μM] 2.5μL 10μL MgCl2 [50mM] 1μL 4μL Miniprep DNA template [2ng/μl] 5μL 20μL Nuclease-free water 27.5μL 110μL iProof DNA Polymerase [2U/μL] 0.5μL 2μL Total volume 50μL 200μL 2.19.5.1 Reaction mix in one Protein LoBind tube for double-digestion of Variable Heavy Chain PCR product Reaction components Volume Purified PCR product [10μg] variable 10x NEBuffer 10μl BSA (100x) 1μl ApaLI [10U/μl] 4μl (40U) NsiI [10U/μl] 6μl (60U) Nuclease-free water Total volume Top up to 100μl 100μl 196 Reaction components Volume Purified PCR product [10μg] variable 10x NEBuffer 10μl BSA (100x) 1μl MfeI [10U/μl] 6μl (60U) XhoI [20U/μl] 3μl (60U) Top up to 100μl Nuclease-free water 100μl Total volume 2.19.5 Reaction mix in one Protein LoBind tube for double-digestion of Variable Light Chain + Constant Lambda Chain PCR product was used. Reaction components Volume Purified PCR product [10μg] variable 10x NEBuffer 10μl BSA (100x) 1μl ApaLI [10U/μl] 5μl (50U) AscI [10U/μl] 5μl (50U) Nuclease-free water Total volume Top up to 100μl 100μl 2.19.6 Reaction mix in one Protein LoBind tube for double-digestion of the vector to allow cloning in the Variable Light Chain fragment 197 Reaction components Volume pDSO IgG1 framework vector [20μg] variable 10x NEBuffer 20μl BSA (100x) 2μl ApaLI [10U/μl] 8μl (80U) PstI [20U/μl] 8μl (160U) Nuclease-free water Total volume Top up to 200μl 200μl 2.19.6 Reaction mix in one Protein LoBind tube for double-digestion of the vector to allow cloning in the Variable Heavy Chain fragment. Reaction components Volume pDSO IgG1 framework vector [20μg] variable 10x NEBuffer 20μl BSA (100x) 2μl MfeI [10U/μl] 10μl (100U) XhoI [20U/μl] 5μl (100U) Nuclease-free water Total volume Top up to 200μl 100μl 2.19.7 Reaction mix for the prevention of self-religation of plasmids. reaction components were placed in a microtube for double-digestion of plasmid Reaction components Volume Double-digested pDSO IgG1 framework vector [20μg] 200μl 10x Antartic Phosphatase reaction buffer 24μl Antartic Phosphatase [5U/μl] Total volume 16μl (80U) 240μl 198 2.19.10 Reaction mix for double-digestion of plasmid constructs containing the Variable Light Chain fragment. Reaction components Volume Master Mix (35 reactions) Miniprep DNA 5.0μl - 10x NEBuffer 1.0μl 35μl BSA (100x) 0.1μl 3.5μl ApaLI [10U/μl] 0.4μl (4U) 14μl PstI [20U/μl] 0.6μl (6U) 21μl Nuclease-free water 2.9μl 101.5μl Total volume 10.0μl Use 5μl/reaction 2.19.10 Reaction mix for double-digestion of plasmid constructs containing the Variable Heavy Chain fragment. Reaction components Volume Master Mix (35 reactions) Miniprep DNA 5.0μl - 10x NEBuffer 1.0μl 35μl BSA (100x) 0.1μl 3.5μl PstI [20U/μl] 0.5μl (4U) 17.5μl XhoI [20U/μl] 0.5μl (6U) 17.5μl Nuclease-free water 2.9μl 101.5μl Total volume 10.0μl Use 5μl/reaction 199 [...]... alter the spatial distances 33 between the glycans on the E proteins, thereby inhibiting the interaction of the virus with DC-SIGN (Pokidysheva, Zhang et al 2006) Another route may involve binding to DIII of the E glycoproteins to prevent binding of the virus to its primary entry receptor (Hung, Hsieh et al 2004) Several studies have shown that the neutralizing epitopes of DV are clustered at the tip of. .. into the Americas correlated with the occurance of DHF/DSS (Rico-Hesse 20 03) and later experiments underline the early replicative advantage of the South-East Asian genotype (Armstrong and Rico-Hesse 20 03; Cologna and Rico-Hesse 20 03; Cologna, Armstrong et al 2005) Dengue virus serotype 3 (DV3) consists of four or five distinct genotypes depending on the analysis performed (Messer, Gubler et al 20 03; ... et al 20 03; Zhang, Chipman et al 20 03) CryoEM has shown the hinge angle between domains I and II of each of the three symmetry-independent E proteins to differ approximately by 5-15° from the crystal structures and about 30 ° from the cryoEM structure of the mature particle (Zhang, Corver et al 20 03) The immature particles formed in the ER mature as they travel through the secretory pathway The slightly... essential for the transcription and translation processes necessary for viral propagation Of the viral NS proteins, the most extensively characterized are the NS3 protein and the cofactor NS2B, and NS5 NS3 protein does not contain long stretches of hydrophobic amino acids but becomes membrane-associated via the interaction with NS2B protein, which together constitutes the functional viral protease, NS2B -3. .. the secretory pathway The slightly acidic pH (~5.8 - 6.0) of the trans Golgi network triggers dissociation of the prM/E heterodimers, which leads to the formation of 90 dimers that lie flat on the surface of the particle, with prM capping the fusion peptide of the E protein This global structural reorganization of the glycoproteins enables the cellular endoprotease furin to cleave prM Furin cleavage... negative-strand template for further generation of the positive strands Since the positive strand serves as both viral genome and mRNA, it is produced in excess of the negative strand The mechanism by which this asymmetric synthesis occurs has yet to be elucidated Cells infected with DV detected at later stages of infection are found to harbor vesicle packets within the cytoplasm of the cells Mackenzie... the generation of membraneassociated M and a pr peptide A recent study has shown that the pr peptide 17 remains associated with the virion until the virus is released to the extracellular milieu to infect neighboring cells (Yu, Zhang et al 2008) (FIG.1.6) FIG.1.6 Life cycle of DV The picture depicts the various cleavage processes within the cell contributing to major conformational changes during the. .. (Falgout, Pethel et al 1991) The N-terminal 180 amino acids of NS3 protein contain the catalytic 14 domain of the viral protease, NS2B -3, as defined by the sequence alignment with known serine proteases of the trypsin superfamily (Bazan and Fletterick 1989), by deletion analyses (Wengler, Czaya et al 1991) and by site-directed mutagenesis of residues in the putative catalytic triad or the substrate-binding... replacement increased the overall abundance of a first serotype but simultaneously decreased the population of the second serotype due to mutations affecting viral fitness In turn, the later decline of the prevalence of the first serotype was associated with the increase in prevalence of the second serotype (Zhang, Mammen et al 2005) Finally, it was suggested that existing clades belonging to the first serotype... dengue genotype and that there is a selection for virulent DV in humans and mosquitoes (Cologna, Armstrong et al 2005) Some dengue genotypes are more capable of producing DHF epidemics in a population base of variable immune status The origin and spread of DHF in the Western Hemisphere can be linked to viruses of the Southeast Asian genotype, whereas the American genotype viruses have been isolated . 6.0) of the trans Golgi network triggers dissociation of the prM/E heterodimers, which leads to the formation of 90 dimers that lie flat on the surface of the particle, with prM capping the. 2001). These data are consistent with the proposed role of NS3 protein in the function of the flaviviral replication complex. The flavivirus NS5 is essential for the capping pathway. NS5 protein. and about 30 ° from the cryoEM structure of the mature particle (Zhang, Corver et al. 20 03) . The immature particles formed in the ER mature as they travel through the secretory pathway. The slightly

Ngày đăng: 11/09/2015, 09:56

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan