Academic literature on the topic 'Avian influenza virus M2e protein'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'Avian influenza virus M2e protein.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "Avian influenza virus M2e protein"

1

Mezhenskaya, Daria, Irina Isakova-Sivak, Victoria Matyushenko, Svetlana Donina, Andrey Rekstin, Konstantin Sivak, Kirill Yakovlev, et al. "Universal Live-Attenuated Influenza Vaccine Candidates Expressing Multiple M2e Epitopes Protect Ferrets against a High-Dose Heterologous Virus Challenge." Viruses 13, no. 7 (June 30, 2021): 1280. http://dx.doi.org/10.3390/v13071280.

Full text
Abstract:
The development of an influenza vaccine with broad protection and durability remains an attractive idea due to the high mutation rate of the influenza virus. An extracellular domain of Matrix 2 protein (M2e) is among the most attractive target for the universal influenza vaccine owing to its high conservancy rate. Here, we generated two recombinant live attenuated influenza vaccine (LAIV) candidates encoding four M2e epitopes representing consensus sequences of human, avian and swine influenza viruses, and studied them in a preclinical ferret model. Both LAIV+4M2e viruses induced higher levels of M2e-specific antibodies compared to the control LAIV strain, with the LAIV/HA+4M2e candidate being significantly more immunogenic than the LAIV/NS+4M2e counterpart. A high-dose heterosubtypic influenza virus challenge revealed the highest degree of protection after immunization with LAIV/HA+4M2e strain, followed by the NS-modified LAIV and the classical LAIV virus. Furthermore, only the immune sera from the LAIV/HA+4M2e-immunized ferrets protected mice from a panel of lethal influenza viruses encoding M genes of various origins. These data suggest that the improved cross-protection of the LAIV/HA+4M2e universal influenza vaccine candidate was mediated by the M2e-targeted antibodies. Taking into account the safety profile and improved cross-protective potential, the LAIV/HA+4M2e vaccine warrants its further evaluation in a phase I clinical trial.
APA, Harvard, Vancouver, ISO, and other styles
2

Shuklina, M. A., L. A. Stepanova, A. A. Kovaleva, A. V. Korotkov, A. A. Shaldzhyan, M. V. Zaitceva, E. I. Eletskaya, and L. M. Tsybalova. "Intranasal immunization with a recombinant protein based on the M2e peptide and second subunit of influenza A viral hemagglutinin fragment induces a cross-protective humoral and Tcell response in mice." Medical Immunology (Russia) 22, no. 2 (April 16, 2020): 357–70. http://dx.doi.org/10.15789/1563-0625-iiw-1584.

Full text
Abstract:
Development of vaccines with a broad-spectrum of protection is one of the priorities in the programs of influenza prevention. Recently, the conserved fragments of influenza virus proteins (M1, M2, NP, the second subunit of the hemagglutinin HA2) provoke interest of investigators as the object of the development a broad-spectrum vaccines. Low immunogenicity present a problem when developing vaccines based on such conserved fragments. However, fusion of low immunogenic antigens into the high immunogenic carrier protein may significantly enhance their immunogenicity. The candidate vaccine protein Flg-HA2-2-4M2e was developed which containins two highly conserved viral antigens (the ectodomain of the M2 protein (M2e), 76130 region of the second subunit of HA2), fused with flagellin as a carrier protein. Flagellin (bacterial flagella protein) is a natural ligand of TLR-5, and has a strong adjuvant activity at different ways of its administration. The purpose of this study was to assess development of humoral and T cell immune response, along with broad-spectrum protection after mice immunization with the candidate Flg-HA2-2-4M2e vaccine protein. Mice were immunized intranasally three times with two-week intervals. Two weeks after the final immunization, the mice were challenged at the 5 LD50 dose with influenza viruses A/California/07/09 (H1N1) pdm09 (phylogenetic group I), or A/Shanghai/2/2013 (H7N9) (phylogenetic group II). The results obtained in this study showed induction of strong M2e-specific humoral response (serum IgG and A) in the immunized mice. Immunization with recombinant protein stimulated formation of M2e-specific and virus-specific CD4+ and CD8+T cells in lung which produced TNFα or IFNγ. Production of antigen-specific effector and central memory T cells was also detected in lungs of immunized mice. The formation of cross-protective immunity in immunized mice was demonstrated in a model of lethal influenza infection. The experimental animals were almost completely protected from the high dose of the pandemic virus A/H1N1pdm09, and highly pathogenic avian influenza A/H7N9 (90-100% survival). We also evaluated the changes of antigen-specific immune response in immunized mice after sublethal infection with A/H3N2 influenza virus. Mice of control and experimental groups were infected with MID100 of influenza virus A/Aichi/2/68 (H3N2). It was shown that the M2e-specific response (IgG, IgA) was significantly increased in immunized mice after sublethal infection with influenza virus A/H3N2, and we detected the changes in profile of M2e-specific IgG subclasses. Following sublethal infection in immunized mice, the proportion of M2e-specific IgG2a was increased 10-fold. The results showed that the recombinant protein Flg-HA2-2-4M2e is a promising candidate for development of universal vaccines, which induces a protective humoral and T-cell response to conserved viral epitopes and protects against influenza A viruses of both phylogenetic groups.
APA, Harvard, Vancouver, ISO, and other styles
3

Kim, Min-Chul, Jun-Gu Choi, Ji-Sun Kwon, Hyun-Mi Kang, Mi-Ra Paek, Ok-Mi Jeong, Jun-Hun Kwon, and Youn-Jeong Lee. "Field Application of the H9M2e Enzyme-Linked Immunosorbent Assay for Differentiation of H9N2 Avian Influenza Virus-Infected Chickens from Vaccinated Chickens." Clinical and Vaccine Immunology 17, no. 12 (October 27, 2010): 1977–84. http://dx.doi.org/10.1128/cvi.00191-10.

Full text
Abstract:
ABSTRACT Vaccination for control of H9N2 low-pathogenicity avian influenza (LPAI) in chickens began in 2007 in South Korea where the H9N2 virus is prevalent. Recently, an enzyme-linked immunosorbent assay (ELISA) using the extracellular domain of the M2 protein (M2e ELISA) was developed as another strategy to differentiate between vaccinated and infected chickens. Here, an ELISA using the extracellular domain of the M2 protein of H9N2 LPAI virus (H9M2e ELISA) was applied to differentiate infected from vaccinated chickens using the H9N2 LPAI virus M2 peptide. The specificity and sensitivity of the optimized H9M2e ELISA were 96.1% and 83.8% (the absorbance of the sample to the absorbance for the positive control [S/P ratio] ≥ 0.6), respectively, with the cutoff value (S/P ratio = 0.6), and the criterion of avian influenza (AI) infection in a chicken house was established as >20% reactivity of anti-M2e antibody per house with this cutoff value. After infection in naïve chickens and once-vaccinated chickens with a hemagglutination inhibition (HI) assay titer of 9.25 ± 0.75 log2 units, the sera from infected chickens were confirmed as AI infected when the chickens were 1 week old in both groups, and AI infection lasted for 24 weeks and 9 weeks in naïve and once-vaccinated chickens, respectively, although in twice-vaccinated chickens with a higher HI titer of 11.17 ± 0.37 log2 units, anti-M2e antibody in infected sera did not reach a level indicating AI infection. In field application, anti-M2e antibody produced in infected chickens after vaccination or in reinfected chickens could be identified as AI infection, although HI test could not distinguish infected from vaccinated sera. These results indicate the utility of H9M2e ELISA as a surveillance tool in control of H9N2 LPAI infections.
APA, Harvard, Vancouver, ISO, and other styles
4

Zhang, Sixin, Xinming Tang, Si Wang, Fangyun Shi, Chunhui Duan, Feifei Bi, Jingxia Suo, et al. "Establishment of Recombinant Eimeria acervulina Expressing Multi-Copies M2e Derived from Avian Influenza Virus H9N2." Vaccines 9, no. 7 (July 16, 2021): 791. http://dx.doi.org/10.3390/vaccines9070791.

Full text
Abstract:
The potential of Eimeria parasites as live vaccine vectors has been reported with successful genetic manipulation on several species like E. tenella, E. mitis and E. necatrix. Among seven Eimeria species infecting chickens, E. acervulina is a highly prevalent, moderately pathogenic species. Thus, it is valuable for the study of transfection and for use as a potential as vaccine vector. In this study, a plasmid containing expression cassette with enhanced yellow fluorescent protein (EYFP), red fluorescent protein (RFP) and 12 copies of extracellular domain of H9N2 avian influenza virus M2 (M2e) protein was used for the transfection. Nucleofected sporozoites were inoculated into birds through wing vein. Recombinant E. acervulina oocysts with 0.1% EYFP+ and RFP+ populations were collected from the feces of the inoculated birds. The fluorescent rate of transgenic parasites reached over 95% after nine successive propagations with a pyrimethamine selection in vivo and fluorescent-activated cell sorting (FACS) of progeny oocysts. The expression of M2e in the transgenic parasites (EaM2e) was confirmed by Western blot and its cytoplasm localization in sporozoites was displayed by an indirect immunofluorescent assay (IFA). Meanwhile, we found that the fecundity of EaM2e was equivalent to that of wild type E. acervulina (EaWT). Taken together, the stable transfection of E. acervulina was successfully established. Future studies will focus on whether transgenic E. acervulina can serve as a live vaccine vector.
APA, Harvard, Vancouver, ISO, and other styles
5

Nemchinov, Lev G., and Angela Natilla. "Transient expression of the ectodomain of matrix protein 2 (M2e) of avian influenza A virus in plants." Protein Expression and Purification 56, no. 2 (December 2007): 153–59. http://dx.doi.org/10.1016/j.pep.2007.05.015.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Park, Ki Seok, Yong Bok Seo, Ji Yeung Lee, Se Jin Im, Sang Hwan Seo, Min Suk Song, Young Ki Choi, and Young Chul Sung. "Complete protection against a H5N2 avian influenza virus by a DNA vaccine expressing a fusion protein of H1N1 HA and M2e." Vaccine 29, no. 33 (July 2011): 5481–87. http://dx.doi.org/10.1016/j.vaccine.2011.05.062.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Swinkels, Willem J. C., Jeroen Hoeboer, Reina Sikkema, Lonneke Vervelde, and Ad P. Koets. "Vaccination induced antibodies to recombinant avian influenza A virus M2 protein or synthetic M2e peptide do not bind to the M2 protein on the virus or virus infected cells." Virology Journal 10, no. 1 (2013): 206. http://dx.doi.org/10.1186/1743-422x-10-206.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Tang, Yinghua, Yuzhen Gong, Yongwei Wang, Peipei Wu, Yamei Liu, Jihu Lu, Feng Gao, Tao Chen, Fengxiang Hou, and Jibo Hou. "Chimaeric VP2 proteins from infectious bursal disease virus containing the N-terminal M2e of H9 subtype avian influenza virus induce neutralizing antibody responses to both viruses." Avian Pathology 42, no. 3 (June 2013): 260–67. http://dx.doi.org/10.1080/03079457.2013.782096.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Reese, Kaleb A., Christopher Lupfer, Rudd C. Johnson, Georgi M. Mitev, Valerie M. Mullen, Bruce L. Geller, and Manoj Pastey. "A Novel Lactococcal Vaccine Expressing a Peptide from the M2 Antigen of H5N2 Highly Pathogenic Avian Influenza A Virus Prolongs Survival of Vaccinated Chickens." Veterinary Medicine International 2013 (2013): 1–8. http://dx.doi.org/10.1155/2013/316926.

Full text
Abstract:
A cost-effective and efficacious influenza vaccine for use in commercial poultry farms would help protect against avian influenza outbreaks. Current influenza vaccines for poultry are expensive and subtype specific, and therefore there is an urgent need to develop a universal avian influenza vaccine. We have constructed a live bacterial vaccine against avian influenza by expressing a conserved peptide from the ectodomain of M2 antigen (M2e) on the surface ofLactococcus lactis(LL). Chickens were vaccinated intranasally with the lactococcal vaccine (LL-M2e) or subcutaneously with keyhole-limpet-hemocyanin conjugated M2e (KLH-M2e). Vaccinated and nonvaccinated birds were challenged with high pathogenic avian influenza virus A subtype H5N2. Birds vaccinated with LL-M2e or KLH-M2e had median survival times of 5.5 and 6.0 days, respectively, which were significantly longer than non-vaccinated birds (3.5 days). Birds vaccinated subcutaneously with KLH-M2e had a lower mean viral burden than either of the other two groups. However, there was a significant correlation between the time of survival and M2e-specific serum IgG. The results of these trials show that birds in both vaccinated groups had significantly (P<0.05) higher median survival times than non-vaccinated birds and that this protection could be due to M2e-specific serum IgG.
APA, Harvard, Vancouver, ISO, and other styles
10

Babapoor, Sankhiros, Tobias Neef, Christian Mittelholzer, Theodore Girshick, Antonio Garmendia, Hongwei Shang, Mazhar I. Khan, and Peter Burkhard. "A Novel Vaccine Using Nanoparticle Platform to Present Immunogenic M2e against Avian Influenza Infection." Influenza Research and Treatment 2011 (January 12, 2011): 1–12. http://dx.doi.org/10.1155/2011/126794.

Full text
Abstract:
Using peptide nanoparticle technology, we have designed two novel vaccine constructs representing M2e in monomeric (Mono-M2e) and tetrameric (Tetra-M2e) forms. Groups of specific pathogen free (SPF) chickens were immunized intramuscularly with Mono-M2e or Tetra-M2e with and without an adjuvant. Two weeks after the second boost, chickens were challenged with 107.2 EID50 of H5N2 low pathogenicity avian influenza (LPAI) virus. M2e-specific antibody responses to each of the vaccine constructs were tested by ELISA. Vaccinated chickens exhibited increased M2e-specific IgG responses for each of the constructs as compared to a non-vaccinated group. However, the vaccine construct Tetra-M2e elicited a significantly higher antibody response when it was used with an adjuvant. On the other hand, virus neutralization assays indicated that immune protection is not by way of neutralizing antibodies. The level of protection was evaluated using quantitative real time PCR at 4, 6, and 8 days post-challenge with H5N2 LPAI by measuring virus shedding from trachea and cloaca. The Tetra-M2e with adjuvant offered statistically significant (P<0.05) protection against subtype H5N2 LPAI by reduction of the AI virus shedding. The results suggest that the self-assembling polypeptide nanoparticle shows promise as a potential platform for a development of a vaccine against AI.
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "Avian influenza virus M2e protein"

1

Tam, Ho-man Alex. "Mechanisms underlying the hyper-induction of tumour necrosis factor alpha (TNF-? by avian influenza virus in human macrophages." Click to view the E-thesis via HKUTO, 2008. http://sunzi.lib.hku.hk/hkuto/record/B41508300.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Tam, Ho-man Alex, and 譚浩文. "Mechanisms underlying the hyper-induction of tumour necrosis factor alpha (TNF-α) by avian influenza virus in human macrophages." Thesis, The University of Hong Kong (Pokfulam, Hong Kong), 2008. http://hub.hku.hk/bib/B41508300.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Hasan, Noor Haliza. "Avian Influenza virus M2e protein: Epitope mapping, competitive ELISA and phage displayed scFv for DIVA in H5N1 serosurveillance." Thesis, 2017. http://hdl.handle.net/2440/119643.

Full text
Abstract:
Within the avian influenza virus (AIV) history, H5N1 subtype is the most alarming in terms of its spread rate throughout the globe with its demonstrated unusual pattern of evolution. Persistency and constant circulation of this subtype in poultry population in a number of countries have resulted its establishment and declaration as enzootic. The affected countries are commonly characterised by high poultry populations and productions. They are also developing countries which have minimal funding allocated for precaution on disease incursion. Past observations showed that a single AIV epizootic is capable of causing significant economic burden throughout the world. Although epizootic, it still resulted sporadic cases of human infection and mortality. Therefore, H5N1 enzootic countries opt for vaccination strategy (usually with inactivated whole virus) to evade AIV incursions. However, this interferes with the AIV surveillance effort. This is due to the lack of diagnostic tool with the ability to differentiate AIV infected animal from vaccinated animal (DIVA). Following this realisation, several options are made available. Diagnostic tool development which is capable of DIVA requires a highly sensitive and specific target which at the same time is economic, and pose ease of application. In recent years, growing interest on the AIV matrix 2 extracellular domain (M2e) protein has propelled its exploration as the target for AIV serosurveillance diagnostic tool development. It has been demonstrated to be highly sensitive and specific in detection for AIV infection in an indirect enzyme-linked immunosorbent assay (ELISA) setting. The factor which made it highly interesting is its ability for DIVA application. M2e protein can only be found in low concentration on an AIV particle which is used in an inactivated vaccination strategy, while present in high concentration if cells are AIV infected. Therefore, this study has further explores the AIV M2e protein potential for AIV serosurveillance diagnostic tool development and successfully demonstrated an M2e-based test in a competitive ELISA format for DIVA. This particular ELISA format was of interest as it can be potentially used in multiple species application, as AIV is a multispecies pathogen. To ensure the universality of the competitor antibody, comparative mapping of anti-M2e antibodies from chicken, mouse and rabbit was done. Findings highlighted slight variations in the epitope identified for the M2e antigen by antibodies from different species. Mouse anti-M2e antibodies are more suitable to be used as the competitor antibodies against anti-M2e chicken sera in the M2e-based competitive ELISA test. Consequently, application of the mouse anti-M2e antibodies in the M2e-based competitive ELISA has demonstrated specific and sensitive indication of AIV infection in the H5N1 challenged chicken sera. Biotechnology developments has also introduced the single chain variable fragment (scFv) antibodies as specific and stable bait for antibodies detection against targeted pathogen’s protein (antigen). Taking advantage of this knowledge, this study has also successfully isolated reactive and specific anti-M2e scFv antibodies from avian sources. This is critical as an avian sourced antibodies to be used as bait for the targeted pathogen’s protein is highly relevant in the setting for AIV serosurveillance application in the poultry industry. These findings are significant in the effort to provide a highly sensitive and specific diagnostic tool, which are also cost effective, easy to apply with high throughput ability. Such ideal diagnostic tool for AIV serosurveillance is highly valuable, as this may hold the key to break the AIV continuous circulation.
Thesis (Ph.D.) -- University of Adelaide, School of Animal and Veterinary Sciences, 2017
APA, Harvard, Vancouver, ISO, and other styles
4

陳羽鴻. "Development of the M2 protein of H1N1 avian influenza virus subunit vaccine." Thesis, 2017. http://ndltd.ncl.edu.tw/handle/bm6f8f.

Full text
Abstract:
碩士
國立嘉義大學
生物農業科技學系研究所
106
Avian influenza(Avian Influenza;AI)is commonly known as avian flu. It is one kind of animal infectious diseases caused by flu viruses. AI viruses can be grouped into highly pathogenic avian influenza (HPAI) and low pathogenic avian influenza (LPAI). The mortality of the poultry infected by HPAI can be higher than 80%. In this study, H1N1 viral M2 gene segment was constructed into the E. coli protein expression vector for expressing the M2 gene 's fusion protein with 8X-Histidine tag. The serum samples of vaccinated mice were assayed by dot blotting and ELISA. The specific antibody against the overexpressed M2 gene's protein produced in the vaccinated mice was determined by dot blotting assay. The M2 gene's fusion protein was also shown to have better immune response in association with the commercial adjuvant. These studies have laid a strong foundation for developing an effective AI subunit vaccine.
APA, Harvard, Vancouver, ISO, and other styles
5

Vuong, Christine. "Evaluation of Sindbis-M2e Virus Vector as a Universal Influenza A Vaccine." Thesis, 2012. http://hdl.handle.net/1969.1/ETD-TAMU-2012-08-11706.

Full text
Abstract:
Although avian influenza virus (AIV) infections in domestic poultry are uncommon, transmission of avian influenza from wild waterfowl reservoirs does occur. Depopulation of the infected flock is the typical response to AIV outbreaks in domestic chicken production, causing a loss in profits and accumulation of unexpected expenses. Because it is impossible to know which of many virus subtypes will cause an outbreak, it is not feasible for the U.S. to stockpile vaccines against all possible avian influenza threats. Currently, the U.S. does not routinely vaccinate chickens against influenza due to the inability to differentiate infected from vaccinated animals (DIVA), which would place limitations on its trade markets. A Sindbis virus vector expressing the PR8 influenza strain's M2e peptide was developed as a potential universal DIVA vaccine. M2e is a conserved peptide amongst influenza A viruses; M2e-specific antibodies induce antibody-dependent cytotoxicity or phagocytosis of infected cells, reducing production and shedding of AIV during infection. In this study, chickens were vaccinated at one-month-of-age with parental (E2S1) or recombinant Sindbis viruses expressing the PR8 M2e peptide (E2S1-M2e) by subcutaneous or intranasal routes at high (106 pfu) or low (103 pfu) dosages. Chickens were boosted at 2-weeks post-initial vaccination using the same virus, route, and dosage, then challenged with low pathogenic H5N3 AIV at 0.2 mL of 106/mL EID50 2-weeks post-boost. Serum samples were collected at 1-week and 2-weeks post-vaccination, 2-weeks post-boost, and 2-weeks post-challenge and screened for PR8 M2e-specific IgY antibody production by ELISA. Both high and low dose subcutaneously, as well as high dose intranasally vaccinated E2S1-M2e groups produced significantly higher levels of PR8 M2e-specific IgY antibodies as early as 1-week post-vaccination, while the uninoculated control and E2S1 groups remained negative for all pre-challenge time points. M2e-specific IgY antibodies capable of binding the challenge H5N3 M2e peptide were detected in groups with existing vaccine-induced M2e-specific antibodies pre-challenge, suggesting antibody M2e cross-reactivity. After challenge, all groups developed M2e-specific IgY antibodies and high HI titers, verifying successful AIV infection during challenge and production of hemagglutinin-specific antibodies. Viral shedding titers 4-days post-challenge were used to measure vaccine efficacy and were similar amongst all groups. Microneutralization assay results confirmed that post-boost serum samples, containing only M2e-specific antibodies, were unable to neutralize AIV in vitro. Although the E2S1-M2e vaccine was capable of producing high levels of M2e-specific IgY antibodies when inoculated subcutaneously, these antibodies were not able to reduce viral shedding and therefore did not protect chickens from AIV.
APA, Harvard, Vancouver, ISO, and other styles
6

Chen, Chien-Hao, and 陳建豪. "Expression and application of avian influenza virus hemagglutinin protein." Thesis, 2006. http://ndltd.ncl.edu.tw/handle/24673044416928532096.

Full text
Abstract:
碩士
國立臺灣大學
獸醫學研究所
94
From 1986 to 2003, the avian influenza viruses (AIV) isolated in Taiwan were all low pathogenic. Some low pathogenic H5N2 AIVs were isolated in Taiwan in 2004, resulting in stamping 380 thousands chickens out from 22 poultry flocks. To test if the virus titers increased during passaging, H5N2 and H6N1 isolates were passaged in specific-pathogen-free embryonated eggs. After 45 passages, the virus titer of H5N2 increased 100 times, and two amino acid residues in HA1 of H5N2 virus were changed. The partial fragments of HA1 relating to neutralizing epitopes of the H5N2 and the H6N1 isolates were cloned in pGEX-5X-1 vector (prokaryotic expression) and pcDNA3.1 vector (eukaryotic expression) for expression. The purified recombinant proteins were intramuscualarly injected in 4-week-old SPF chickens for two times. Two weeks after the second immunization, the serum from H6-immunized chickens showed hemagglutination inhibition (HI) titers of 24 to 26, but no antibody was detected in H5-immunized chickens. To test the increase efficiency of liposome compounds on vaccine, Newcastle disease vaccines coated with liposome and with liposome/chitosan were used to immunize chickens. The results showed that Newcastle disease vaccine coated with liposome/chitosan increased HI titers in vaccinated chickens but not vaccine coated with lipsome alone.
APA, Harvard, Vancouver, ISO, and other styles
7

Luo, Yu-Li, and 羅昱立. "Studies of avian influenza virus-induced apoptosis and HA protein expression." Thesis, 2006. http://ndltd.ncl.edu.tw/handle/28070314762861980293.

Full text
Abstract:
碩士
國立屏東科技大學
生物科技研究所
94
Avian influenza virus(AIV)belongs to orthomyxoviridae family. Highly pathogenetic avian influenza virus causes flowl plague in chickens and turkeys. The HA protein of AIV 1209 strain was expressed by Pichia pastoris yeast expression system. The results indicated that the tagged fusion protein was present both in medium and pellet. The HA protein could be cleaved into HA1(47 kDa) and HA2(22 kDa). The AIV CE9 could induce apoptosis in MDCK cell. To further study the molecular mechanism of AIV-induced apoptosis, the NA protein of AIV H5N2 CE9 strain was cloned into pcDNA3.1(-), and the recombinant DNA transfected into MDCK cell lines. The results indicated that at the same focus, caspase 3 were not detected in the MDCK cell expressing the GFP-NA fusion protein. Phylogenesis analysis of HA and NA gene of strains Taiwanese 1209 and CE9 was done to sequence with other H5N2 viruses in the world using MEGA 3.1 software. The result indicate the 1209 and CE9 strain belong to the same group as the Italy strain.
APA, Harvard, Vancouver, ISO, and other styles
8

Singh, Shailbala. "First Characterization of Avian Memory T Lymphocyte Responses to Avian Influenza Virus Proteins." 2009. http://hdl.handle.net/1969.1/ETD-TAMU-2009-12-7534.

Full text
Abstract:
Although wild birds are natural hosts of avian influenza viruses (AIVs), these viruses can be highly contagious to poultry and a zoonotic threat to humans. The propensity of AIV for genetic variation through genetic shift and drift allows virus to evade vaccine mediated humoral immunity. An alternative approach to current vaccine development is induction of CD8+ T cells which responds to more conserved epitopes than humoral immunity and targets a broader spectrum of viruses. Since the memory CD8+ T lymphocyte responses in chickens to individual AIV proteins have not been defined, the modulation of responses of the memory CD8+ T lymphocytes to H5N9 AIV hemagglutinin (HA) and nucleocapsid (NP) proteins over a time course were evaluated. CD8+ T lymphocyte responses induced by intramuscular inoculation of chickens with AIV HA and NP expressing cDNA plasmids or a non-replicating human adenovirus vector were identified through ex vivo stimulation with virus infected, major histocompatibility complex (MHC) matched antigen presenting cells (APCs). The IFN? production by activated lymphocytes was evaluated by macrophage production of nitric oxide and ELISA. MHC-I restricted memory T lymphocyte responses were determined at 10 days and 3, 5, 7 and 9 weeks post-inoculation (p.i). The use of non-professional APCs and APC driven proliferation of cells with CD8+ phenotype correlated with the activation of CD8+ T lymphocytes. The responses specific to nucleocapsid protein (NP) were consistently greater than those to the hemagglutinin (HA) at 5 weeks when the CD8+ T cell responses were maximum. By 8 to 9 weeks p.i., responses to either protein were undetectable. The T lymphocytes also responded to stimulation with a heterologous H7N2 AIV infected APCs. Administration of booster dose induced secondary effector cell mediated immune responses which had greater magnitudes than primary effector responses at 10 days p.i. Flow cytometric analysis (FACS) of the T lymphocytes demonstrated that memory CD8+ T lymphocytes of chickens can be distinguished from naive lymphocytes by their higher expression of CD44 and CD45 surface antigens. CD45 expression of memory lymphocytes further increases upon ex vivo stimulation with APCs expressing AIV. This is the first characterization of avian memory responses following both primary and secondary expression of any individual viral protein.
APA, Harvard, Vancouver, ISO, and other styles
9

Chien, Pin-Cheng, and 錢品澄. "Characterization of Monoclonal Antibodies against Nonstructural Protein 1 (NS1) of Avian Influenza Virus." Thesis, 2014. http://ndltd.ncl.edu.tw/handle/43455856100074850441.

Full text
Abstract:
碩士
國立臺灣大學
獸醫學研究所
102
NS1 is an indictor Ag of influenza virus (IV) infection and it’s only produced during IV replication in very early stage of infection. It can be used in determining whether the chicken was infected or not, which also can be applied on early diagnosis of IV infection. Therefore, my study aimed to characterize the anti-NS1 MAb (αNS1 MAb) by indirect ELISA (iELISA), indirect immunofluorescence assay (IFA), western blotting (WB), immunohistochemistry stain (IHC) and mapping the antibody binding site by eukaryotic expression system (EES). Subsequently, to develop a rapid, sensitive and specific diagnostic MAbs-based NS1 Ag sandwich ELISA that might be incorporated with NP and M Ag ELISA kit for detecting the IV infection. The parental hybridoma have been prepared by immunizing Balb/c mice with E. coli expressed recombinant nonstructural protein 1 (rNS1) oringinated from IV isolates of A/chicken/Taiwan/2838V/00 (H6N1/2838). 16 αNS1 MAbs have been characterized by WB, IFA and iELISA with NS1 Ag from several IV isolates such as A/chicken/Miaoli/2904/00 (H6N1/2904), A/chicken/Taiwan/1209/03 (H5N2/1209). Meanwhile, the epitope mapping was studied in EES. Expression of full length rNS1 (residues 1-230) were all positive and 4 αNS1 MAb (4M4, 4M6, 4N5, 4R3) were negative in C-terminal deletion (residues 1-207). Accroding to epitope mapping analysis results, which can divide 16 αNS1 MAb into four groups (A, B, C &; D). The predictive epitopes of 16 αNS1 MAb mainly recognize the effector domain (residues 74-230) and group B recognize the C-terminal tail (residues 202-230) of NS1 protein. MAb-based NS1 Ag sandwich ELISA was designed as sixteen MAb individually conjugated with HRP as the tracer Ab paired with each non-conjugated MAb acted as docking Ab respectively to compare the binding ability of those combinations with E. coli expressed rNS1. Current results indicated that MP-5 (4R11 – 4M2/HRP), MP-9 (4R11 – 4J12/HRP) and MP-10 (4M2 – 4M2/HRP) can detect NS1 Ag from 15 subtypes of IV (H1~H15). It is believed the three MP (MP-5, MP-9 &; MP-10) we studied have potential to be applied on detecting the IV NS1 Ag.
APA, Harvard, Vancouver, ISO, and other styles
10

陳廷軒. "Immunogenicity of recombinant protein, adenovirus vector and virus-like particles of the avian-origin influenza H7N9 virus hemagglutinin." Thesis, 2014. http://ndltd.ncl.edu.tw/handle/7ed3h7.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Books on the topic "Avian influenza virus M2e protein"

1

Alexander, D. J., N. Phin, and M. Zuckerman. Influenza. Edited by I. H. Brown. Oxford University Press, 2011. http://dx.doi.org/10.1093/med/9780198570028.003.0037.

Full text
Abstract:
Influenza is a highly infectious, acute illness which has affected humans and animals since ancient times. Influenza viruses form the Orthomyxoviridae family and are grouped into types A, B, and C on the basis of the antigenic nature of the internal nucleocapsid or the matrix protein. Infl uenza A viruses infect a large variety of animal species, including humans, pigs, horses, sea mammals, and birds, occasionally producing devastating pandemics in humans, such as in 1918 when it has been estimated that between 50–100 million deaths occurred worldwide.There are two important viral surface glycoproteins, the haemagglutinin (HA) and neuraminidase (NA). The HA binds to sialic acid receptors on the membrane of host cells and is the primary antigen against which a host’s antibody response is targeted. The NA cleaves the sialic acid bond attaching new viral particles to the cell membrane of host cells allowing their release. The NA is also the target of the neuraminidase inhibitor class of antiviral agents that include oseltamivir and zanamivir and newer agents such as peramivir. Both these glycoproteins are important antigens for inducing protective immunity in the host and therefore show the greatest variation.Influenza A viruses are classified into 16 antigenically distinct HA (H1–16) and 9 NA subtypes (N1–9). Although viruses of relatively few subtype combinations have been isolated from mammalian species, all subtypes, in most combinations, have been isolated from birds. Each virus possesses one HA and one NA subtype.Last century, the sudden emergence of antigenically different strains in humans, termed antigenic shift, occurred on three occasions, 1918 (H1N1), 1957 (H2N2) and 1968 (H3N2), resulting in pandemics. The frequent epidemics that occur between the pandemics are as a result of gradual antigenic change in the prevalent virus, termed antigenic drift. Epidemics throughout the world occur in the human population due to infection with influenza A viruses, such as H1N1 and H3N2 subtypes, or with influenza B virus. Phylogenetic studies have led to the suggestion that aquatic birds that show no signs of disease could be the source of many influenza A viruses in other species. The 1918 H1N1 pandemic strain is thought to have arisen as a result of spontaneous mutations within an avian H1N1 virus. However, most pandemic strains, such as the 1957 H2N2, 1968 H3N2 and 2009 pandemic H1N1, are considered to have emerged by genetic re-assortment of the segmented RNA genome of the virus, with the avian and human influenza A viruses infecting the same host.Influenza viruses do not pass readily between humans and birds but transmission between humans and other animals has been demonstrated. This has led to the suggestion that the proposed reassortment of human and avian influenza viruses takes place in an intermediate animal with subsequent infection of the human population. Pigs have been considered the leading contender for the role of intermediary because they may serve as hosts for productive infections of both avian and human viruses, and there is good evidence that they have been involved in interspecies transmission of influenza viruses; particularly the spread of H1N1 viruses to humans. Apart from public health measures related to the rapid identification of cases and isolation. The main control measures for influenza virus infections in human populations involves immunization and antiviral prophylaxis or treatment.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography