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

To see the other types of publications on this topic, follow the link: Avian influenza virus M2e protein.

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

Select a source type:

Consult the top 50 journal articles for your research 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.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

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
11

Cho, Ki Joon, Bert Schepens, Jong Hyeon Seok, Sella Kim, Kenny Roose, Ji-Hye Lee, Rodrigo Gallardo, et al. "Structure of the Extracellular Domain of Matrix Protein 2 of Influenza A Virus in Complex with a Protective Monoclonal Antibody." Journal of Virology 89, no. 7 (January 21, 2015): 3700–3711. http://dx.doi.org/10.1128/jvi.02576-14.

Full text
Abstract:
ABSTRACTThe extracellular domain of influenza A virus matrix protein 2 (M2e) is conserved and is being evaluated as a quasiuniversal influenza A vaccine candidate. We describe the crystal structure at 1.6 Å resolution of M2e in complex with the Fab fragment of an M2e-specific monoclonal antibody that protects against influenza A virus challenge. This antibody binds M2 expressed on the surfaces of cells infected with influenza A virus. Five out of six complementary determining regions interact with M2e, and three highly conserved M2e residues are critical for this interaction. In this complex, M2e adopts a compact U-shaped conformation stabilized in the center by the highly conserved tryptophan residue in M2e. This is the first description of the three-dimensional structure of M2e.IMPORTANCEM2e of influenza A is under investigation as a universal influenza A vaccine, but its three-dimensional structure is unknown. We describe the structure of M2e stabilized with an M2e-specific monoclonal antibody that recognizes natural M2. We found that the conserved tryptophan is positioned in the center of the U-shaped structure of M2e and stabilizes its conformation. The structure also explains why previously reportedin vivoescape viruses, selected with a similar monoclonal antibody, carried proline residue substitutions at position 10 in M2.
APA, Harvard, Vancouver, ISO, and other styles
12

Wang, Bao-Zhong, Harvinder S. Gill, Sang-Moo Kang, Li Wang, Ying-Chun Wang, Elena V. Vassilieva, and Richard W. Compans. "Enhanced Influenza Virus-Like Particle Vaccines Containing the Extracellular Domain of Matrix Protein 2 and a Toll-Like Receptor Ligand." Clinical and Vaccine Immunology 19, no. 8 (May 30, 2012): 1119–25. http://dx.doi.org/10.1128/cvi.00153-12.

Full text
Abstract:
ABSTRACTThe extracellular domain of matrix protein 2 (M2e) is conserved among influenza A viruses. The goal of this project is to develop enhanced influenza vaccines with broad protective efficacy using the M2e antigen. We designed a membrane-anchored fusion protein by replacing the hyperimmunogenic region ofSalmonella entericaserovar Typhimurium flagellin (FliC) with four repeats of M2e (4.M2e-tFliC) and fusing it to a membrane anchor from influenza virus hemagglutinin (HA). The fusion protein was incorporated into influenza virus M1-based virus-like particles (VLPs). These VLPs retained Toll-like receptor 5 (TLR5) agonist activity comparable to that of soluble FliC. Mice immunized with the VLPs by either intramuscular or intranasal immunization showed high levels of systemic M2-specific antibody responses compared to the responses to soluble 4.M2e protein. High mucosal antibody titers were also induced in intranasally immunized mice. All intranasally immunized mice survived lethal challenges with live virus, while intramuscularly immunized mice showed only partial protection, revealing better protection by the intranasal route. These results indicate that a combination of M2e antigens and TLR ligand adjuvants in VLPs has potential for development of a broadly protective influenza A virus vaccine.
APA, Harvard, Vancouver, ISO, and other styles
13

Heinen, Paul P., Frans A. Rijsewijk, Els A. de Boer-Luijtze, and André T. J. Bianchi. "Vaccination of pigs with a DNA construct expressing an influenza virus M2–nucleoprotein fusion protein exacerbates disease after challenge with influenza A virus." Journal of General Virology 83, no. 8 (August 1, 2002): 1851–59. http://dx.doi.org/10.1099/0022-1317-83-8-1851.

Full text
Abstract:
In mice, vaccines inducing antibodies to the extracellular domain of the M2 protein (M2e) can confer protection to influenza A virus infection. Unlike the surface glycoproteins, haemagglutinin and neuraminidase, this domain of M2 is highly conserved and is therefore a potential broad-spectrum immunogen. In this study, the protection conferred by vaccines inducing antibodies to M2e was evaluated in a challenge model for swine influenza in pigs. A protein resulting from the fusion between M2e and the hepatitis B virus core protein (M2eHBc), with or without adjuvant, was evaluated. In addition, a DNA construct expressing a fusion protein between M2e and influenza virus nucleoprotein (M2eNP) was evaluated to see if the broad-spectrum protection conferred by antibodies could be further enhanced by T helper cells and cytotoxic T cells. All vaccines induced an antibody response against M2e, and the M2eNP DNA vaccine additionally induced an influenza virus-specific lymphoproliferation response. However, after challenge with a swine influenza virus (H1N1), no protection was observed in the vaccinated groups compared with the non-vaccinated control group. On the contrary, vaccinated pigs showed more severe clinical signs than the control pigs. The M2eNP DNA-vaccinated pigs showed the most severe clinical signs and three out of six pigs died on days 1 and 2 post-challenge. These results indicate that antibodies to M2e, especially in combination with cell-mediated immune responses, exacerbate disease. Thus, clinical signs after infection should be observed closely in further studies using M2e as an immunogen and caution should be exercised in using M2e in humans.
APA, Harvard, Vancouver, ISO, and other styles
14

Zharikova, Darya, Krystyna Mozdzanowska, Jingqi Feng, Manxin Zhang, and Walter Gerhard. "Influenza Type A Virus Escape Mutants Emerge In Vivo in the Presence of Antibodies to the Ectodomain of Matrix Protein 2." Journal of Virology 79, no. 11 (June 1, 2005): 6644–54. http://dx.doi.org/10.1128/jvi.79.11.6644-6654.2005.

Full text
Abstract:
ABSTRACT The ectodomain of matrix protein 2 (M2e) of human influenza type A virus strains has remained remarkably conserved since 1918. Because M2e-specific immunity has been shown to decrease morbidity and mortality associated with influenza virus infection in several animal models and because natural infection and current vaccines do not appear to induce a good M2e-specific antibody (Ab) response, M2e has been considered as potential vaccine for inducing cross-reactive protection against influenza type A viruses. The high degree of structural conservation of M2e could in part be the consequence of a poor M2e-specific Ab response and thus the absence of pressure for change. To assess this possibility, we studied the course of infection in SCID mice in the presence or absence of passive M2e-specific monoclonal Abs (MAbs). We found that virus mutants with antigenic changes in M2e emerged in 65% of virus-infected mice treated with M2e-specific but not control MAbs. However, the diversity of escape mutants was highly restricted since only two types were isolated from 22 mice, one with a proline-to-leucine and the other with a proline-to-histidine interchange at amino acid position 10 of M2e. The implications of these findings for the use of M2e as a broadly protective vaccine are discussed.
APA, Harvard, Vancouver, ISO, and other styles
15

Braz Gomes, Kimberly, Sucheta D’Sa, Grace Lovia Allotey-Babington, Sang-Moo Kang, and Martin J. D’Souza. "Transdermal Vaccination with the Matrix-2 Protein Virus-like Particle (M2e VLP) Induces Immunity in Mice against Influenza A Virus." Vaccines 9, no. 11 (November 15, 2021): 1324. http://dx.doi.org/10.3390/vaccines9111324.

Full text
Abstract:
In this study, our goal was to utilize the extracellular domain matrix-2 protein virus-like particle (M2e VLP) that has been found to be highly conserved amongst all strains of influenza and could serve as a potential vaccine candidate against influenza. Previous studies have demonstrated that the VLP of the M2e showed increased activation of innate and adaptive immune responses. Therefore, to further explore its level of efficacy and protection, this vaccine was administered transdermally and tested in a pre-clinical mouse model. The M2e VLP was encapsulated into a polymeric matrix with the addition of Alhydrogel® and Monophosphoryl Lipid-A (MPL-A®), together referred to as AS04. The M2e VLP formulations induced IgG titers, with increased levels of IgG1 in the M2e VLP MP groups and further elevated levels of IgG2a were found specifically in the M2e VLP MP Adjuvant group. This trend in humoral immunity was also observed from a cell-mediated standpoint, where M2e VLP MP groups showed increased expression in CD4+ T cells in the spleen and the lymph node and high levels of CD8+ T cells in the lymph node. Taken together, the results illustrate the immunogenic potential of the matrix-2 protein virus-like particle (M2e VLP) vaccine.
APA, Harvard, Vancouver, ISO, and other styles
16

Fiers, Walter>, Sabine Neirynck, Tom Deroo, Xavier Saelens, and Willy Min Jou. "Soluble recombinant influenza vaccines." Philosophical Transactions of the Royal Society of London. Series B: Biological Sciences 356, no. 1416 (December 29, 2001): 1961–63. http://dx.doi.org/10.1098/rstb.2001.0980.

Full text
Abstract:
Soluble, recombinant forms of influenza A virus haemagglutinin and neuraminidase have been produced in cells of lower eukaryotes, and shown in a mouse model to induce complete protective immunity against a lethal virus challenge. Soluble neuraminidase, produced in a baculovirus system, consisted of tetramers, dimers and monomers. Only the tetramers were enzymatically active. The immunogenicity decreased very considerably in the order tetra > di > mono. Therefore, we fused the head part of the neuraminidase gene to a tetramerizing leucine zipper sequence; the resulting product was enzymatically active, tetrameric neuraminidase. The protective immunity induced by this engineered neuraminidase, however, remained fairly strain–specific. A third influenza A virus protein, the M2 protein, has only 23 amino acids exposed on the outer membrane surface. This extracellular part, M2e, has been remarkably conserved in all human influenza A strains since 1933. By fusing the M2e sequence to hepatitis B virus core protein, we could obtain highly immunogenic particles that induced complete, strain–independent, long–lasting protection in mice against a lethal viral challenge. Native M2 is a tetrameric protein and this conformation of the M2e part can also be mimicked by fusing this sequence to a tetramerizing leucine zipper. The potential of the resulting protein as a vaccine candidate remains to be evaluated.
APA, Harvard, Vancouver, ISO, and other styles
17

Ingrole, Rohan S., Wenqian Tao, Jatindra N. Tripathy, and Harvinder S. Gill. "Synthesis and Immunogenicity Assessment of Elastin-Like Polypeptide-M2e Construct as an Influenza Antigen." Nano LIFE 04, no. 02 (June 2014): 1450004. http://dx.doi.org/10.1142/s1793984414500044.

Full text
Abstract:
The 23 amino acid-long extracellular domain of the influenza virus transmembrane protein M2 (M2e) has remained highly conserved since the 1918 pandemic, and is thus considered a good candidate for development of a universal influenza A vaccine. However, M2e is poorly immunogenic. In this study we assessed the potential of increasing immunogenicity of M2e by constructing a nanoscale-designed protein polymer containing the M2e sequence and an elastin-like polypeptide (ELP) nanodomain consisting of alanine and tyrosine guest residues (ELP(A2YA2)24). The ELP nanodomain was included to increase antigen size, and to exploit the inherent thermal inverse phase transition behavior of ELPs to purify the protein polymer. The ELP(A2YA2)24 + M2e nanodomained molecule was recombinantly synthesized. Characterization of its inverse phase transition behavior demonstrated that attachment of M2e to ELP(A2YA2)24 increased its transition temperature compared to ELP(A2YA2)24. Using a dot blot test we determined that M2e conjugated to ELP is recognizable by M2e-specific antibodies, suggesting that the conjugation process does not adversely affect the immunogenic property of M2e. Further, upon vaccinating mice with ELP(A2YA2)24 + M2e it was found that indeed the nanodomained protein enhanced M2e-specific antibodies in mouse serum compared to free M2e peptide and ELP(A2YA2)24. The immune serum could also recognize M2 expressed on influenza virions. Overall, this data suggests the potential of using molecules containing M2e-ELP nanodomains to develop a universal influenza vaccine.
APA, Harvard, Vancouver, ISO, and other styles
18

Tsybalova, L. M., L. A. Stepanova, R. Yu Kotlyarov, E. A. Blokhina, M. A. Shuklina, E. S. Mardanova, A. V. Korotkov, M. V. Potapchuk, and N. V. Ravin. "Strengthening the Effectiveness of the Candidate Influenza Vaccine by Combining Conserved Sequences of Hemagglutinin and M2 protein." Epidemiology and Vaccine Prevention 16, no. 3 (June 20, 2017): 65–70. http://dx.doi.org/10.31631/2073-3046-2017-16-3-65-70.

Full text
Abstract:
The development of universal influenza vaccine - a vaccine directed to all subtypes of human influenza A viruses - is the really actual problem task. This paper presents the comparative characteristic of the specific activity of various recombinant proteins consisting of antigenic determinants of influenza A virus - the ectodomain of the M2 protein (M2e) and a fragment of the second subunit of the hemagglutinin (the amino acid sequence 76 - 130). Flagellin - Salmonella typhimurium protein was used as carrier protein and as adjuvant. We use two forms of flagellin: full size and with deleted hypervariable region. The proteins showed high immunogenicity, and the ability to prevent lethal infection of influenza virus in mice. Full-length flagellin with HA2 (76 - 130) and M2e on the C-terminus (protein Flg-HA2-4M2e) demonstrated the most protective properties. It provides 100% survival immunized mice that were challenge with a high dose of influenza A (H3N2) - 10 LD50. Proteins containing only full sized flagellin with M2e or flagellin truncated form with M2e at the C-terminus and HA2 within the hypervariable region, protected 75% of animals from lethal infection. Protein Flg-HA2-4M2e is promising for further study as a vaccine.
APA, Harvard, Vancouver, ISO, and other styles
19

Wang, Li, Ying-Chun Wang, Hao Feng, Tamanna Ahmed, Richard W. Compans, and Bao-Zhong Wang. "Virus-Like Particles Containing the Tetrameric Ectodomain of Influenza Matrix Protein 2 and Flagellin Induce Heterosubtypic Protection in Mice." BioMed Research International 2013 (2013): 1–12. http://dx.doi.org/10.1155/2013/686549.

Full text
Abstract:
The ectodomain of matrix protein 2 (M2e) is highly conserved among influenza A viruses and can be a promising candidate antigen for a broadly cross-protective vaccine. In this study, a tetrameric M2e (tM2e) and a truncated form of flagellin (tFliC) were coincorporated into virus-like particles (VLPs) to enhance its immunogenicity. Our data showed that the majority of M2e in VLPs was presented as tetramers by introducing a foreign tetramerization motif GCN4. Intranasal immunization with tM2e VLPs significantly enhanced the levels of serum IgG and IgG subclasses compared to soluble M2e (sM2e) in mice. tM2e VLPs also induced higher M2e-specific T-cell and mucosal antibody responses, conferring complete protection against homologous influenza virus infection. The immunogenicity of tM2e VLPs was further enhanced by coincorporation of the membrane-anchored tFliC (tM2e chimeric VLPs) or coadministration with tFliC VLPs as a mixture, but not the soluble flagellin, inducing strong humoral and cellular immune responses conferring cross-protection against lethal challenge with heterotypic influenza viruses. These results support the development of tM2e chimeric VLPs as universal vaccines and warrant further investigation.
APA, Harvard, Vancouver, ISO, and other styles
20

Zykova, Anna A., Elena A. Blokhina, Roman Y. Kotlyarov, Liudmila A. Stepanova, Liudmila M. Tsybalova, Victor V. Kuprianov, and Nikolai V. Ravin. "Highly Immunogenic Nanoparticles Based on a Fusion Protein Comprising the M2e of Influenza A Virus and a Lipopeptide." Viruses 12, no. 10 (October 6, 2020): 1133. http://dx.doi.org/10.3390/v12101133.

Full text
Abstract:
The highly conserved extracellular domain of the transmembrane protein M2 (M2e) of the influenza A virus is a promising target for the development of broad-spectrum vaccines. However, M2e is a poor immunogen by itself and must be linked to an appropriate carrier to induce an efficient immune response. In this study, we obtained recombinant mosaic proteins containing tandem copies of M2e fused to a lipopeptide from Neisseria meningitidis surface lipoprotein Ag473 and alpha-helical linkers and analyzed their immunogenicity. Six fusion proteins, comprising four or eight tandem copies of M2e flanked by alpha-helical linkers, lipopeptides, or a combination of both of these elements, were produced in Escherichia coli. The proteins, containing both alpha-helical linkers and lipopeptides at each side of M2e repeats, formed nanosized particles, but no particulate structures were observed in the absence of lipopeptides. Animal study results showed that proteins with lipopeptides induced strong M2e-specific antibody responses in the absence of external adjuvants compared to similar proteins without lipopeptides. Thus, the recombinant M2e-based proteins containing alpha-helical linkers and N. meningitidis lipopeptide sequences at the N- and C-termini of four or eight tandem copies of M2e peptide are promising vaccine candidates.
APA, Harvard, Vancouver, ISO, and other styles
21

Sun, Weina, Allen Zheng, Robert Miller, Florian Krammer, and Peter Palese. "An Inactivated Influenza Virus Vaccine Approach to Targeting the Conserved Hemagglutinin Stalk and M2e Domains." Vaccines 7, no. 3 (September 18, 2019): 117. http://dx.doi.org/10.3390/vaccines7030117.

Full text
Abstract:
Universal influenza virus vaccine candidates that focus on the conserved hemagglutinin (HA) stalk domain and the extracellular domain of the matrix protein 2 (M2e) have been developed to increase the breadth of protection against multiple strains. In this study, we report a novel inactivated influenza virus vaccine approach that combines these two strategies. We inserted a human consensus M2e epitope into the immunodominant antigenic site (Ca2 site) of three different chimeric HAs (cHAs). Sequential immunization with inactivated viruses containing these modified cHAs substantially enhanced M2e antibody responses while simultaneously boosting stalk antibody responses. The combination of additional M2e antibodies with HA stalk antibodies resulted in superior antibody-mediated protection in mice against challenge viruses expressing homologous or heterosubtypic hemagglutinin and neuraminidase compared to vaccination strategies that targeted the HA stalk or M2e epitopes in isolation.
APA, Harvard, Vancouver, ISO, and other styles
22

Blokhina, Elena A., Eugenia S. Mardanova, Liudmila A. Stepanova, Liudmila M. Tsybalova, and Nikolai V. Ravin. "Plant-Produced Recombinant Influenza A Virus Candidate Vaccine Based on Flagellin Linked to Conservative Fragments of M2 Protein and Hemagglutintin." Plants 9, no. 2 (January 29, 2020): 162. http://dx.doi.org/10.3390/plants9020162.

Full text
Abstract:
The development of recombinant influenza vaccines with broad spectrum protection is an important task. The combination of conservative viral antigens, such as M2e, the extracellular domain of the transmembrane protein M2, and conserved regions of the second subunit of hemagglutinin (HA), provides an opportunity for the development of universal influenza vaccines. Immunogenicity of the antigens could be enhanced by fusion to bacterial flagellin, the ligand for Toll-like receptor 5, acting as a powerful mucosal adjuvant. In this study, we report the transient expression in plants of a recombinant protein comprising flagellin of Salmonella typhimurium fused to the conserved region of the second subunit of HA (76–130 a.a.) of the first phylogenetic group of influenza A viruses and four tandem copies of the M2e peptide. The hybrid protein was expressed in Nicotiana benthamiana plants using the self-replicating potato virus X-based vector pEff up to 300 µg/g of fresh leaf tissue. The intranasal immunization of mice with purified fusion protein induced high levels of M2e-specific serum antibodies and provided protection against lethal challenge with influenza A virus strain A/Aichi/2/68(H3N2). Our results show that M2e and hemagglutinin-derived peptide can be used as important targets for the development of a plant-produced vaccine against influenza.
APA, Harvard, Vancouver, ISO, and other styles
23

Samal, Sweety, Tripti Shrivastava, Praveen Sonkusre, Zaigham Abbas Rizvi, Rajesh Kumar, Shubbir Ahmed, Preeti Vishwakarma, et al. "Tetramerizing tGCN4 domain facilitates production of Influenza A H1N1 M2e higher order soluble oligomers that show enhanced immunogenicity in vivo." Journal of Biological Chemistry 295, no. 42 (August 13, 2020): 14352–66. http://dx.doi.org/10.1074/jbc.ra120.013233.

Full text
Abstract:
One strategy for the development of a next generation influenza vaccine centers upon using conserved domains of the virus to induce broader and long-lasting immune responses. The production of artificial proteins by mimicking native-like structures has shown to be a promising approach for vaccine design against diverse enveloped viruses. The amino terminus of influenza A virus matrix 2 ectodomain (M2e) is highly conserved among influenza subtypes, and previous studies have shown M2e-based vaccines are strongly immunogenic, making it an attractive target for further exploration. We hypothesized that stabilizing M2e protein in the mammalian system might influence the immunogenicity of M2e with the added advantage to robustly produce the large scale of proteins with native-like fold and hence can act as an efficient vaccine candidate. In this study, we created an engineered construct in which the amino terminus of M2e is linked to the tetramerizing domain tGCN4, expressed the construct in a mammalian system, and tested for immunogenicity in BALB/c mice. We have also constructed a stand-alone M2e construct (without tGCN4) and compared the protein expressed in mammalian cells and in Escherichia coli using in vitro and in vivo methods. The mammalian-expressed protein was found to be more stable, more antigenic than the E. coli protein, and form higher-order oligomers. In an intramuscular protein priming and boosting regimen in mice, these proteins induced high titers of antibodies and elicited a mixed Th1/Th2 response. These results highlight the mammalian-expressed M2e soluble proteins as a promising vaccine development platform.
APA, Harvard, Vancouver, ISO, and other styles
24

Mytle, Nutan, Sonja Leyrer, Jon R. Inglefield, Andrea M. Harris, Thomas E. Hickey, Jacob Minang, Hang Lu, et al. "Influenza Antigens NP and M2 Confer Cross Protection to BALB/c Mice against Lethal Challenge with H1N1, Pandemic H1N1 or H5N1 Influenza A Viruses." Viruses 13, no. 9 (August 27, 2021): 1708. http://dx.doi.org/10.3390/v13091708.

Full text
Abstract:
Influenza hemagglutinin (HA) is considered a major protective antigen of seasonal influenza vaccine but antigenic drift of HA necessitates annual immunizations using new circulating HA versions. Low variation found within conserved non-HA influenza virus (INFV) antigens may maintain protection with less frequent immunizations. Conserved antigens of influenza A virus (INFV A) that can generate cross protection against multiple INFV strains were evaluated in BALB/c mice using modified Vaccinia virus Ankara (MVA)-vectored vaccines that expressed INFV A antigens hemagglutinin (HA), matrix protein 1 (M1), nucleoprotein (NP), matrix protein 2 (M2), repeats of the external portion of M2 (M2e) or as tandem repeats (METR), and M2e with transmembrane region and cytoplasmic loop (M2eTML). Protection by combinations of non-HA antigens was equivalent to that of subtype-matched HA. Combinations of NP and forms of M2e generated serum antibody responses and protected mice against lethal INFV A challenge using PR8, pandemic H1N1 A/Mexico/4108/2009 (pH1N1) or H5N1 A/Vietnam/1203/2004 (H5N1) viruses, as demonstrated by reduced lung viral burden and protection against weight loss. The highest levels of protection were obtained with NP and M2e antigens delivered as MVA inserts, resulting in broadly protective immunity in mice and enhancement of previous natural immunity to INFV A.
APA, Harvard, Vancouver, ISO, and other styles
25

Hasan, Noor Haliza, Esmaeil Ebrahimie, Jagoda Ignjatovic, Simson Tarigan, Anne Peaston, and Farhid Hemmatzadeh. "Epitope Mapping of Avian Influenza M2e Protein: Different Species Recognise Various Epitopes." PLOS ONE 11, no. 6 (June 30, 2016): e0156418. http://dx.doi.org/10.1371/journal.pone.0156418.

Full text
APA, Harvard, Vancouver, ISO, and other styles
26

Mezhenskaya, Daria, Irina Isakova-Sivak, Tatiana Gupalova, Elena Bormotova, Eugenia Kuleshevich, Tatiana Kramskaya, Galina Leontieva, Larisa Rudenko, and Alexander Suvorov. "A Live Probiotic Vaccine Prototype Based on Conserved Influenza a Virus Antigens Protect Mice against Lethal Influenza Virus Infection." Biomedicines 9, no. 11 (October 21, 2021): 1515. http://dx.doi.org/10.3390/biomedicines9111515.

Full text
Abstract:
Background: Due to the highly variable nature of the antigenic properties of the influenza virus, many efforts have been made to develop broadly reactive influenza vaccines. Various vaccine platforms have been explored to deliver conserved viral antigens to the target cells to induce cross-reactive immune responses. Here, we assessed the feasibility of using Enterococcus faecium L3 as a bacterial vector for oral immunization against influenza virus. Methods: we generated two vaccine prototypes by inserting full-length HA2 (L3-HA2) protein or its long alpha helix (LAH) domain in combination with four M2e tandem repeats (L3-LAH+M2e) into genome of E.faecium L3 probiotic strain. The immunogenicity and protective potential of these oral vaccines were assessed in a lethal challenge model in BALB/c mice. Results: as expected, both vaccine prototypes induced HA stem-targeting antibodies, whereas only L3-LAH+4M2e vaccine induced M2e-specific antibody. The L3-HA2 vaccine partially protected mice against lethal challenge with two H1N1 heterologous viruses, while 100% of animals in the L3-LAH+4M2e vaccine group survived in both challenge experiments, and there was significant protection against weight loss in this group, compared to the L3 vector-immunized control mice. Conclusions: the recombinant enterococcal strain L3-LAH+4M2e can be considered as a promising live probiotic vaccine candidate for influenza prevention and warrants further evaluation in relevant pre-clinical models.
APA, Harvard, Vancouver, ISO, and other styles
27

Cho, Ki Joon, Bert Schepens, Kristof Moonens, Lei Deng, Walter Fiers, Han Remaut, and Xavier Saelens. "Crystal Structure of the Conserved Amino Terminus of the Extracellular Domain of Matrix Protein 2 of Influenza A Virus Gripped by an Antibody." Journal of Virology 90, no. 1 (October 14, 2015): 611–15. http://dx.doi.org/10.1128/jvi.02105-15.

Full text
Abstract:
We report the crystal structure of the M2 ectodomain (M2e) in complex with a monoclonal antibody that binds the amino terminus of M2. M2e extends into the antibody binding site to form an N-terminal β-turn near the bottom of the paratope. This M2e folding differs significantly from that of M2e in complex with an antibody that binds another part of M2e. This suggests that M2e can adopt at least two conformations that can elicit protective antibodies.
APA, Harvard, Vancouver, ISO, and other styles
28

Song, Manki, and Byoung-Shik Shim. "Sublingual immunization with M2-based vaccine induces broad protective immunity (P4301)." Journal of Immunology 190, no. 1_Supplement (May 1, 2013): 123.14. http://dx.doi.org/10.4049/jimmunol.190.supp.123.14.

Full text
Abstract:
Abstract Background: M2e-based vaccine constructs have been shown to be more protective when administered by the intranasal (i.n.) route than after parenteral injection. However, i.n. administration of vaccines poses rare but serious safety issues associated with retrograde passage of inhaled antigens and adjuvants through the olfactory epithelium. In this study we examined whether the sublingual (s.l.) route could serve as a safe and effective alternative mucosal delivery route for administering a prototype M2e-based vaccine. The mechanism whereby s.l. immunization with M2e vaccine candidate induces broad protection against infection with different influenza virus subtypes was explored. Methods and Results: A recombinant M2 protein with three tandem copies of the M2e (3M2eC) was expressed in Escherichia coli. Parenteral immunizations of mice with 3M2eC induced high levels of M2e-specific serum Abs but failed to provide complete protection against lethal challenge with influenza virus. In contrast, s.l. immunization with 3M2eC was superior for inducing protection in mice. In the latter animals, protection was associated with specific Ab responses in the lungs. Conclusions: The results demonstrate that s.l. immunization with 3M2eC vaccine induced airway mucosal immune responses along with broad cross-protective immunity to influenza. These findings may contribute to the understanding of the M2-based vaccine approach to control epidemic and pandemic influenza infections.
APA, Harvard, Vancouver, ISO, and other styles
29

Ninyio, Nathaniel Nyakaat, Kok Lian Ho, Abdul Rahman Omar, Wen Siang Tan, Munir Iqbal, and Abdul Razak Mariatulqabtiah. "Virus-like Particle Vaccines: A Prospective Panacea Against an Avian Influenza Panzootic." Vaccines 8, no. 4 (November 19, 2020): 694. http://dx.doi.org/10.3390/vaccines8040694.

Full text
Abstract:
Epizootics of highly pathogenic avian influenza (HPAI) have resulted in the deaths of millions of birds leading to huge financial losses to the poultry industry worldwide. The roles of migratory wild birds in the harbouring, mutation, and transmission of avian influenza viruses (AIVs), and the lack of broad-spectrum prophylactic vaccines present imminent threats of a global panzootic. To prevent this, control measures that include effective AIV surveillance programmes, treatment regimens, and universal vaccines are being developed and analysed for their effectiveness. We reviewed the epidemiology of AIVs with regards to past avian influenza (AI) outbreaks in birds. The AIV surveillance programmes in wild and domestic birds, as well as their roles in AI control were also evaluated. We discussed the limitations of the currently used AI vaccines, which necessitated the development of a universal vaccine. We evaluated the current development of AI vaccines based upon virus-like particles (VLPs), particularly those displaying the matrix-2 ectodomain (M2e) peptide. Finally, we highlighted the prospects of these VLP vaccines as universal vaccines with the potential of preventing an AI panzootic.
APA, Harvard, Vancouver, ISO, and other styles
30

Kwak, Chaewon, Quyen Thi Nguyen, and Haryoung Poo. "Fusion protein (M2e-HA2-NP) adjuvanted with γ-PGA/Alum induce the cross-reactivity against heterologous influenza A viruses." Journal of Immunology 204, no. 1_Supplement (May 1, 2020): 245.10. http://dx.doi.org/10.4049/jimmunol.204.supp.245.10.

Full text
Abstract:
Abstract The epidemic and pandemic influenza viruses result in substantial morbidity and mortality in humans. Vaccination is the most effective way to prevent influenza virus infection. In this study, we cloned the conserved regions of influenza A virus, extracellular domain of matrix protein 2 (M2e), hemagglutinin 2 (HA2) and nucleoprotein (NP). Then, we generated fusion (M2e-HA2-NP) protein as a universal vaccine antigen candidate. γ-PGA/Alum (PA) was used as an adjuvant to enhance the efficacy of fusion protein. In mice challenged with influenza viruses, fusion protein (15 μg) provided 100% protection against A/Puerto Rico/8/1934 (H1N1; PR8) or A/California/04/09 (pH1N1; CA04) and 80% protection against H3N2 (a reassortant virus carrying HA and NA genes of A/Hong Kong/1/68). In dose-sparing effect experiments, low dose (1 or 5 μg) of fusion protein with PA provided the same protection comparable to 15 μg of fusion protein against H1N1, pH1N1 and H3N2. Humoral and cellular immunities against heterologous influenza viruses were significantly enhanced in mice vaccinated with fusion protein mixed with PA compared with alum-adjuvanted fusion protein. Taken together, the fusion protein adjuvanted with PA may be a good vaccine candidate showing heterosubtypic cross-reactivity.
APA, Harvard, Vancouver, ISO, and other styles
31

Chang, Kevin W., Eugene V. Barsov, Andrea L. Ferris, and Stephen H. Hughes. "Mutations of a Residue within the Polyproline-Rich Region of Env Alter the Replication Rate and Level of Cytopathic Effects in Chimeric Avian Retroviral Vectors." Journal of Virology 79, no. 16 (August 15, 2005): 10258–67. http://dx.doi.org/10.1128/jvi.79.16.10258-10267.2005.

Full text
Abstract:
ABSTRACT Previous attempts to extend the host range of the avian sarcoma/leukosis virus (ASLV)-based RCASBP vectors produced two viral vectors, RCASBP M2C (4070A) and RCASBP M2C (797-8), which replicate using the amphotropic murine leukemia virus 4070A Env protein (2). Both viruses were adapted to replicate efficiently in the avian cell line DF-1, but RCASBP M2C (4070A) caused extensive cytopathic effects (CPE) in DF-1 cells whereas RCASBP M2C (797-8) induced low levels of CPE. The two viruses differed only at amino acid 242 of the polyproline-rich region in the surface (SU) subunit of the Env protein. In RCASBP M2C (4070A), an isoleucine replaced the wild-type proline residue, whereas a threonine residue was found in RCASBP M2C (797-8). In the present study, we show that other amino acid substitutions at position 242 strongly influence the CPE and replication rate of the chimeric viruses. There was a correlation between the amount of unintegrated linear retroviral DNA present in infected DF-1 cells and the level of CPE. This suggests that there may be a role for superinfection in the CPE. The treatment of RCASBP M2C (4070A)-infected cells with dantrolene, which inhibits the release of calcium from the endoplasmic reticulum (ER), reduced the amount of CPE seen during infection with the highly cytotoxic virus. Dantrolene treatment did not appear to affect virus production, suggesting that Ca2+ release from the ER had a role in the CPE caused by these viruses.
APA, Harvard, Vancouver, ISO, and other styles
32

Stepanova, L. A., R. Y. Kotlyarov, M. A. Shuklina, E. A. Blochina, M. V. Sergeeva, M. V. Potapchuk, A. A. Kovaleva, N. V. Ravin, and L. M. Tsybalova. "Influence of the Linking Order of Fragments of HA2 and M2e of the influenza A Virus to Flagellin on the Properties of Recombinant Proteins." Acta Naturae 10, no. 1 (March 15, 2018): 85–94. http://dx.doi.org/10.32607/20758251-2018-10-1-85-94.

Full text
Abstract:
The ectodomain of the M2 protein (M2e) and the conserved fragment of the second subunit of hemagglutinin (HA2) are promising candidates for broadly protective vaccines. In this paper, we report on the design of chimeric constructs with differing orders of linkage of four tandem copies of M2e and the conserved fragment of HA2 (76-130) from phylogenetic group II influenza A viruses to the C-terminus of flagellin. The 3D-structure of two chimeric proteins showed that interior location of the M2e tandem copies (Flg-4M2e-HA2) provides partial -helix formation nontypical of native M2e on the virion surface. The C-terminal position of the M2e tandem copies (Flg-HA2-4M2e) largely retained its native M2e conformation. These conformational differences in the structure of the two chimeric proteins were shown to affect their immunogenic properties. Different antibody levels induced by the chimeric proteins were detected. The protein Flg-HA2-4M2e was more immunogenic as compared to Flg-4M2e-HA2, with the former offering full protection to mice against a lethal challenge. We obtained evidence suggesting that the order of linkage of target antigens in a fusion protein may influence the 3D conformation of the chimeric construct, which leads to changes in immunogenicity and protective potency.
APA, Harvard, Vancouver, ISO, and other styles
33

Kirsteina, Anna, Inara Akopjana, Janis Bogans, Ilva Lieknina, Juris Jansons, Dace Skrastina, Tatjana Kazaka, et al. "Construction and Immunogenicity of a Novel Multivalent Vaccine Prototype Based on Conserved Influenza Virus Antigens." Vaccines 8, no. 2 (April 24, 2020): 197. http://dx.doi.org/10.3390/vaccines8020197.

Full text
Abstract:
Influenza, an acute, highly contagious respiratory disease, remains a significant threat to public health. More effective vaccination strategies aimed at inducing broad cross-protection not only against seasonal influenza variants, but also zoonotic and emerging pandemic influenza strains are urgently needed. A number of conserved protein targets to elicit such cross-protective immunity have been under investigation, with long alpha-helix (LAH) from hemagglutinin stalk and ectodomain of matrix protein 2 ion channel (M2e) being the most studied ones. Recently, we have reported the three-dimensional structure and some practical applications of LAH expressed in Escherichia coli system (referred to as tri-stalk protein). In the present study, we investigated the immunogenicity and efficacy of a panel of broadly protective influenza vaccine prototypes based on both influenza tri-stalk and triple M2e (3M2e) antigens integrated into phage AP205 virus-like particles (VLPs). While VLPs containing the 3M2e alone induced protection against standard homologous and heterologous virus challenge in mice, only the combination of both conserved influenza antigens into a single VLP fully protected mice from a high-dose homologous H1N1 influenza infection. We propose that a combination of genetic fusion and chemical coupling techniques to expose two different foreign influenza antigens on a single particle is a perspective approach for generation of a broadly-effective vaccine candidate that could protect against the constantly emerging influenza virus strains.
APA, Harvard, Vancouver, ISO, and other styles
34

Subroto, Toto, Ari Hardianto, Abdul Alim Kahari, and Tika Pradnjaparamita. "Sintesis Tiga Peptida Bergugus Pelindung sebagai Prekursor Komponen Vaksin Influenza Universal." Jurnal Natur Indonesia 15, no. 2 (July 27, 2015): 84. http://dx.doi.org/10.31258/jnat.15.2.84-91.

Full text
Abstract:
Current highly effective conventional vaccine to halt the spread of bird flu has not been invented yet because of susceptiblemutation of influenza virus. In spite of undergoing mutation which causes the amino acid sequence change, influenzaviruses maintain conservation at ectodomain of M2 protein, especially M2e(2-16) (SLLTEVETPIRNEW). The use ofconserved epitope M2e(2-16) in epitope-based vaccine potentially produces universal influenza vaccine. In designingepitope-based vaccine, the M2e(2-16) needs to be coupled with T helper epitope, P25, which is subsequently mentioned asM2e(2-16)-K-P25 (SLLTEVETPIRNEWGKKKL IPNASLIENCTKAEL). The M2e(2-16)-K-P25 was synthesized usingconvergent solid phase peptide synthesis strategy because of the size of the sequence. In this strategy, four peptideprecursors of M2e(2-16)-K-P25; SLLTEVETP (F1), IRNEWGK (F2), KLIPNASLI (F3), and ENCTKAEL (F4); were synthesizedin advance. After the precursors ready, coupling reaction was performed to obtain M2e(2-16)-K-P25. In the previousresearch, F3 has been obtained in high purity through Fmoc/tBu solid phase peptide synthesis method. In this conductedresearch, the three remaining precursors; F1, F2, and F4; were synthesized by the same method. Each peptide was analysedby thin layer chromatography, HPLC, and mass spectroscopy methods. F1, F2 and F4 were successfully synthesized andeach of them was detected at 1490.0, 1874.8 and 1881.9 amu, respectively. However, F1 was not possible to purify becauseof its insolubility in various solvents.
APA, Harvard, Vancouver, ISO, and other styles
35

Blokhina, E. A., and N. V. Ravin. "CONSTRUCTION OF MOSAIC HBC PARTICLES PRESENTING CONSERVATIVE FRAGMENTS OF M2 PROTEIN AND HEMAGGLUTININ OF INFLUENZA A VIRUS." Problems of Virology, Russian journal 63, no. 3 (June 20, 2018): 130–35. http://dx.doi.org/10.18821/0507-4088-2018-63-3-130-135.

Full text
Abstract:
Virus-like HBc particles formed as a result of the self-assembly of the nuclear antigen of the hepatitis B virus can be used as a highly immunogenic carrier for the presentation of foreign epitopes when creating recombinant vaccines. We use this vehicle to create influenza vaccines based on the conservative antigens of the influenza virus, the extracellular domain of the transmembrane protein M2 (M2e) and the fragment of the second subunit of hemagglutinin (HA2). Presentation on the surface of HBc particles should improve the immunogenicity of these peptides. Using genetic engineering techniques, we obtained a fusion protein in which the HA2 sequence is attached to the N-terminus of the HBc antigen, and the M2e peptide is included in the immunodominant loop region exposed on the surface of HBc particle. The hybrid protein expressed in Escherichia coli and purified under denaturing conditions formed virus-like HBc particles after refolding in vitro. Refolding of this protein in the presence of a previously denatured HBc antigen carrying no inserts resulted in formation of mosaic virus-like particles. The developed method will allow construction of mosaic HBc particles carrying different target epitopes of the influenza virus by combining the corresponding modified HBc proteins, which opens the possibility of creating vaccines with a wider spectrum of protection.
APA, Harvard, Vancouver, ISO, and other styles
36

Ong, Hui Kian, Chean Yeah Yong, Wen Siang Tan, Swee Keong Yeap, Abdul Rahman Omar, Mariatulqabtiah Abdul Razak, and Kok Lian Ho. "An Influenza A Vaccine Based on the Extracellular Domain of Matrix 2 Protein Protects BALB/C Mice Against H1N1 and H3N2." Vaccines 7, no. 3 (August 19, 2019): 91. http://dx.doi.org/10.3390/vaccines7030091.

Full text
Abstract:
Current seasonal influenza A virus (IAV) vaccines are strain-specific and require annual reconstitution to accommodate the viral mutations. Mismatches between the vaccines and circulating strains often lead to high morbidity. Hence, development of a universal influenza A vaccine targeting all IAV strains is urgently needed. In the present study, the protective efficacy and immune responses induced by the extracellular domain of Matrix 2 protein (M2e) displayed on the virus-like particles of Macrobrachium rosenbergii nodavirus (NvC-M2ex3) were investigated in BALB/c mice. NvC-M2ex3 was demonstrated to be highly immunogenic even in the absence of adjuvants. Higher anti-M2e antibody titers corresponded well with increased survival, reduced immunopathology, and morbidity of the infected BALB/c mice. The mice immunized with NvC-M2ex3 exhibited lower H1N1 and H3N2 virus replication in the respiratory tract and the vaccine activated the production of different antiviral cytokines when they were challenged with H1N1 and H3N2. Collectively, these results suggest that NvC-M2ex3 could be a potential universal influenza A vaccine.
APA, Harvard, Vancouver, ISO, and other styles
37

Firsov, Aleksey, Irina Tarasenko, Tatiana Mitiouchkina, Natalya Ismailova, Lyubov Shaloiko, Alexander Vainstein, and Sergey Dolgov. "High-Yield Expression of M2e Peptide of Avian Influenza Virus H5N1 in Transgenic Duckweed Plants." Molecular Biotechnology 57, no. 7 (March 5, 2015): 653–61. http://dx.doi.org/10.1007/s12033-015-9855-4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
38

Mezhenskaya, Daria, Irina Isakova-Sivak, Tatiana Kotomina, Victoria Matyushenko, Min-Chul Kim, Noopur Bhatnagar, Ki-Hye Kim, Sang-Moo Kang, and Larisa Rudenko. "A Strategy to Elicit M2e-Specific Antibodies Using a Recombinant H7N9 Live Attenuated Influenza Vaccine Expressing Multiple M2e Tandem Repeats." Biomedicines 9, no. 2 (February 1, 2021): 133. http://dx.doi.org/10.3390/biomedicines9020133.

Full text
Abstract:
Influenza viruses remain a serious public health problem. Vaccination is the most effective way to prevent the disease; however, seasonal influenza vaccines demonstrate low or no effectiveness against antigenically drifted and newly emerged influenza viruses. Different strategies of eliciting immune responses against conserved parts of various influenza virus proteins are being developed worldwide. We constructed a universal live attenuated influenza vaccine (LAIV) candidate with enhanced breadth of protection by modifying H7N9 LAIV by incorporating four epitopes of M2 protein extracellular part into its hemagglutinin molecule. The new recombinant H7N9+4M2e vaccine induced anti-M2e antibody responses and demonstrated increased protection against heterosubtypic challenge viruses in direct and serum passive protection studies, compared to the classical H7N9 LAIV. The results of our study suggest that the H7N9+4M2e warrants further investigation in pre-clinical and phase 1 clinical trials.
APA, Harvard, Vancouver, ISO, and other styles
39

Mardanova, Eugenia S., and Nikolai V. Ravin. "Plant-produced Recombinant Influenza A Vaccines Based on the M2e Peptide." Current Pharmaceutical Design 24, no. 12 (July 5, 2018): 1317–24. http://dx.doi.org/10.2174/1381612824666180309125344.

Full text
Abstract:
Background: Influenza is a widely distributed infection that almost annually causes seasonal epidemics. The current egg-based platforms for influenza vaccine production are facing a number of challenges and are failing to satisfy the global demand in the case of pandemics due to the long production time. Recombinant vaccines are an alternative that can be quickly produced in high quantities in standard expression systems. Methods: : Plants may become a promising biofactory for the large-scale production of recombinant proteins due to low cost, scalability, and safety. Plant-based expression systems have been used to produce recombinant vaccines against influenza based on two targets; the major surface antigen hemagglutinin and the transmembrane protein M2. <P> Results: Different forms of recombinant hemagglutinin were successfully expressed in plants, and some plantproduced vaccines based on hemagglutinin were successfully tested in clinical trials. However, these vaccines remain strain specific, while the highly conserved extracellular domain of the M2 protein (M2e) could be used for the development of a universal influenza vaccine. In this review, the state of the art in developing plant-produced influenza vaccines based on M2e is presented and placed in perspective. A number of strategies to produce M2e in an immunogenic form in plants have been reported, including its presentation on the surface of plant viruses or virus-like particles formed by capsid proteins, linkage to bacterial flagellin, and targeting to protein bodies. Conclusion: Some M2e-based vaccine candidates were produced at high levels (up to 1 mg/g of fresh plant tissue) and were shown to be capable of stimulating broad-range protective immunity.
APA, Harvard, Vancouver, ISO, and other styles
40

Kim, Ki-Hye, Young-Man Kwon, Young-Tae Lee, Min-Chul Kim, Hye Hwang, Eun-Ju Ko, Youri Lee, Hyo-Jick Choi, and Sang-Moo Kang. "Virus-Like Particles Are a Superior Platform for Presenting M2e Epitopes to Prime Humoral and Cellular Immunity against Influenza Virus." Vaccines 6, no. 4 (September 20, 2018): 66. http://dx.doi.org/10.3390/vaccines6040066.

Full text
Abstract:
Influenza virus M2 protein has a highly conserved ectodomain (M2e) as a cross-protective antigenic target. We investigated the antigenic and immunogenic properties of tandem repeat M2e (5xM2e) proteins and virus-like particles (5xM2e VLP) to better understand how VLP and protein platform vaccines induce innate and protective adaptive immune responses. Despite the high antigenic properties of 5xM2e proteins, the 5xM2e VLP was superior to 5xM2e proteins in inducing IgG2a isotype antibodies, T cell responses, plasma cells and germinal center B cells as well as in conferring cross protection. Mice primed with 5xM2e VLP were found to be highly responsive to 5xM2e protein boost, overcoming the low immunogenicity and protective efficacy of 5xM2e proteins. Immunogenic differences between VLPs and proteins in priming immune responses might be due to an intrinsic ability of 5xM2e VLP to stimulate dendritic cells secreting T helper type 1 (Th1) cytokines. We also found that 5xM2e VLP was effective in inducing inflammatory cytokines and chemokines, and in recruiting macrophages, monocytes, neutrophils, and CD11b+ dendritic cells at the injection site. Therefore, this study provides evidence that 5xM2e VLP is an effective vaccine platform, inducing cross-protection by stimulating innate and adaptive immune responses.
APA, Harvard, Vancouver, ISO, and other styles
41

Sumarningsih, Sumarningsih, Simson Tarigan, H. Farhid, and Jagoda Ignjatovic. "Characterisation of M2e Antigenicity using anti-M2 Monoclonal Antibody and anti-M2e Polyclonal Antibodies." Jurnal Ilmu Ternak dan Veteriner 24, no. 3 (September 24, 2019): 122. http://dx.doi.org/10.14334/jitv.v24i3.1987.

Full text
Abstract:
Matrix 2 ectodomain (M2e) protein is a potential antigen for detection of influenza A virus infection in vaccinated poultry (DIVA test). However the M2e antigenicity and immune response it induces in either humans or animals are poorly understood. Seventeen M2e peptides and sixteen recombinant M2e (rM2e) proteins with amino acid (aa) changes introduced at position 10, 11, 12, 13 14, 16, 18 and 20 were compared by western blot (WB) and enzyme-linked immunosorbent assay (ELISA) using mouse anti-M2 monoclonal antibody (mAb) 14C2, and anti-M2e peptide chicken and rabbit polyclonal antibody (pAb). The mAb 14C had the best discriminating power and indicated that all six positions contributed to the M2e antigenicity. Position 11 was the important immunodominant and affected Mab14C binding to a greatest degree. Changes in the adjacent position 14, 16 and 18 also influenced the binding, and it detected regardless of the method (WB or ELISA), or the antigen used (M2e peptide or rM2e). For chicken pAb and rabbit pAb, the immunodominant aa was position 10 and the antibody reaction was not affected by aa change at 11. The binding of rabbit pAb was also affected by changes at 14 and 16, which confirm the contribution of these positions to the M2e antigenicity. Position 10 was the only important position for the binding of chicken pAb to M2e. Overall, the study showed that the M2e antigenic sites are located between residues 10 – 18 and that aa changes at position 10, 11, 12, 14, 16 and 18 may all affect the antibody binding within the M2e protein.
APA, Harvard, Vancouver, ISO, and other styles
42

Zahmanova, Gergana G., Milena Mazalovska, Katerina H. Takova, Valentina T. Toneva, Ivan N. Minkov, Eugenia S. Mardanova, Nikolai V. Ravin, and George P. Lomonossoff. "Rapid High-Yield Transient Expression of Swine Hepatitis E ORF2 Capsid Proteins in Nicotiana benthamiana Plants and Production of Chimeric Hepatitis E Virus-Like Particles Bearing the M2e Influenza Epitope." Plants 9, no. 1 (December 24, 2019): 29. http://dx.doi.org/10.3390/plants9010029.

Full text
Abstract:
The Hepatitis E virus (HEV) is a causative agent of acute hepatitis, mainly transmitted by the fecal-oral route or zoonotic. Open reading frame (ORF) 2 encodes the viral capsid protein, which is essential for virion assembly, host interaction, and inducing neutralizing antibodies. In this study, we investigated whether full-length and N- and C-terminally modified versions of the capsid protein transiently expressed in N. benthamiana plants could assemble into highly-immunogenic, virus-like particles (VLPs). We also assessed whether such VLPs can act as a carrier of foreign immunogenic epitopes, such as the highly-conserved M2e peptide from the Influenza virus. Plant codon-optimized HEV ORF2 capsid genes were constructed in which the nucleotides coding the N-terminal, the C-terminal, or both parts of the protein were deleted. The M2e peptide was inserted into the P2 loop after the residue Gly556 of HEV ORF2 protein by gene fusion, and three different chimeric constructs were designed. Plants expressed all versions of the HEV capsid protein up to 10% of total soluble protein (TSP), including the chimeras, but only the capsid protein consisting of aa residues 110 to 610 (HEV 110–610) and chimeric M2 HEV 110–610 spontaneously assembled in higher order structures. The chimeric VLPs assembled into particles with 22–36 nm in diameter and specifically reacted with the anti-M2e antibody.
APA, Harvard, Vancouver, ISO, and other styles
43

Schotsaert, Michael, Tine Ysenbaert, Anouk Smet, Bert Schepens, Walter Fiers, and Xavier Saelens. "Infection-permissive immunity provided by NA- and M2e-based vaccines protects against influenza A virus challenge and allows the induction of heterosubtypic immunity during subsequent infections. (VAC5P.1127)." Journal of Immunology 194, no. 1_Supplement (May 1, 2015): 73.12. http://dx.doi.org/10.4049/jimmunol.194.supp.73.12.

Full text
Abstract:
Abstract Licensed influenza vaccines protect by inducing neutralizing antibodies against the main viral surface antigens hemagglutinin (HA) and neuraminidase (NA). Influenza infection induces an antiviral T-cell response that correlates with protection against a subsequent infection with a virus of a different subtype (heterosubtypic immunity, HSI). Sterilizing immunity provided by classical vaccines however, may impair the induction of cellular HSI. We previously demonstrated in mice that vaccination with virus like particles containing the ectodomain of Matrix 2 protein (M2e-VLP) induce infection-permissive immunity that not only protects against influenza A viruses of different subtypes, but also allows the induction of cellular HSI. We could recently show that infection-permissive immunity provided by vaccination with recombinantly produced soluble tetrameric N1 NA, alone or combined with M2e-VLP, not only is protective against homologous challenge with pandemic 2009 H1N1 virus but also allows establishment of cellular HSI that protects against challenge with H3N2 virus. NA-immune animals also have an enhanced frequency of circulating functional antiviral memory CD8+ T-cells upon rechallenge with heterologous virus. In contrast, vaccination with recombinant soluble trimeric H1 HA derived from pandemic 2009 H1N1 virus induces strain-specific sterilizing immunity without infection-induced cellular HSI and fails to protect against challenge with H3N2 virus.
APA, Harvard, Vancouver, ISO, and other styles
44

Zinyakov, N. G., A. V. Andriyasov, Ye V. Ovchinnikova, A. A. Kozlov, P. D. Zhestkov, D. B. Andreychuk, and I. A. Chvala. "Analysis of marker substitutions in A/chicken/Astrakhan/2171-1/2020 H5N8 isolate of avian influenza virus recovered in the Astrakhan Oblast." Veterinary Science Today, no. 2 (July 1, 2021): 132–37. http://dx.doi.org/10.29326/2304-196x-2021-2-37-132-137.

Full text
Abstract:
At the end of 2020, a large-scale bird death was registered at one of the poultry farms in the Astrakhan region, the cause of which was avian influenza. Data on detection of the marker substitutions in viral proteins of avian influenza virus A/chicken/Astrakhan/2171-1/2020 isolate are presented in the paper. Type A Н5N8 avian influenza virus was identified with complex PCR-based methods in the submitted samples. Hemagglutinin gene fragment sequencing identified REKRRKR/ GLF, highly pathogenic avian influenza virus isolate-characteristic amino acid sequence of the hemagglutinin cleavage site. Phylogenetic analysis of nucleotide sequences of hemagglutinin gene segment (848–1105 bp ORF) allowed A/chicken/Astrakhan/2171-1/2020 H5N8 isolate to be classified to highly pathogenic avian influenza virus genetic clade 2.3.4.4. Comparative analysis of genome segments using available databases showed that A/chicken/Astrakhan/2171-1/2020 H5N8 virus related to А/Н5 avian influenza virus isolates detected in the Russian Federation in 2016–2020. Analysis of the studied virus isolate hemagglutinin amino acid identified AIV-characteristic G225QRG228 amino acids in the receptor-binding domain of the protein enabling high-affinity binding to avian epithelial cell SAα-2,3- gal receptors. Single mutations, 70G in NEP protein and 13Р in PB1 protein, out of the list of the reported influenza virus mutations affecting successful influenza virus replication in mammals were identified. No mutations affecting virus sensitivity to anti-viral medicines, rimantadin, amantadine, oseltamivir and zanamivir, were detected. The following mutations recognized as pathogenicity determinants in mice were found: 42S in the NS1 protein and 30D protein 215A in M1 protein.
APA, Harvard, Vancouver, ISO, and other styles
45

Tarasenko, I. V., A. I. Taranov, A. P. Firsov, and S. V. Dolgov. "Expression of the nucleotide sequence for the M2e peptide of avian influenza virus in transgenic tobacco plants." Applied Biochemistry and Microbiology 49, no. 8 (November 8, 2013): 695–701. http://dx.doi.org/10.1134/s0003683813080061.

Full text
APA, Harvard, Vancouver, ISO, and other styles
46

Golchin, Mehdi, Masoud Moghadaszadeh, Hadi Tavakkoli, Reza Ghanbarpour, and Siavoush Dastmalchi. "Recombinant M2e-HA2 fusion protein induced immunity responses against intranasally administered H9N2 influenza virus." Microbial Pathogenesis 115 (February 2018): 183–88. http://dx.doi.org/10.1016/j.micpath.2017.12.050.

Full text
APA, Harvard, Vancouver, ISO, and other styles
47

Scholtissek, Christoph, Jürgen Stech, Scott Krauss, and Robert G. Webster. "Cooperation between the Hemagglutinin of Avian Viruses and the Matrix Protein of Human Influenza A Viruses." Journal of Virology 76, no. 4 (February 15, 2002): 1781–86. http://dx.doi.org/10.1128/jvi.76.4.1781-1786.2002.

Full text
Abstract:
ABSTRACT To analyze the compatibility of avian influenza A virus hemagglutinins (HAs) and human influenza A virus matrix (M) proteins M1 and M2, we doubly infected Madin-Darby canine kidney cells with amantadine (1-aminoadamantane hydrochloride)-resistant human viruses and amantadine-sensitive avian strains. By using antisera against the human virus HAs and amantadine, we selected reassortants containing the human virus M gene and the avian virus HA gene. In our system, high virus yields and large, well-defined plaques indicated that the avian HAs and the human M gene products could cooperate effectively; low virus yields and small, turbid plaques indicated that cooperation was poor. The M gene products are among the primary components that determine the species specificities of influenza A viruses. Therefore, our system also indicated whether the avian HA genes effectively reassorted into the genome and replaced the HA gene of the prevailing human influenza A viruses. Most of the avian HAs that we tested efficiently cooperated with the M gene products of the early human A/PR/8/34 (H1N1) virus; however, the avian HAs did not effectively cooperate with the most recently isolated human virus that we tested, A/Nanchang/933/95 (H3N2). Cooperation between the avian HAs and the M proteins of the human A/Singapore/57 (H2N2) virus was moderate. These results suggest that the currently prevailing human influenza A viruses might have lost their ability to undergo antigenic shift and therefore are unable to form new pandemic viruses that contain an avian HA, a finding that is of great interest for pandemic planning.
APA, Harvard, Vancouver, ISO, and other styles
48

Tarigan, Simson, Risa Indriani, Peter A. Durr, and Jagoda Ignjatovic. "Characterization of the M2e antibody response following highly pathogenic H5N1 avian influenza virus infection and reliability of M2e ELISA for identifying infected among vaccinated chickens." Avian Pathology 44, no. 4 (July 4, 2015): 259–68. http://dx.doi.org/10.1080/03079457.2015.1042428.

Full text
APA, Harvard, Vancouver, ISO, and other styles
49

Kim, Eun-Ha, Jun-Han Lee, Philippe Noriel Q. Pascua, Min-Suk Song, Yun-Hee Baek, Hyeok-il Kwon, Su-Jin Park, et al. "Prokaryote-expressed M2e protein improves H9N2 influenza vaccine efficacy and protection against lethal influenza a virus in mice." Virology Journal 10, no. 1 (2013): 104. http://dx.doi.org/10.1186/1743-422x-10-104.

Full text
APA, Harvard, Vancouver, ISO, and other styles
50

Ghorbani, Amir, John M. Ngunjiri, Ming Xia, Mohamed Elaish, Hyesun Jang, K. C. Mahesh, Michael C. Abundo, Xi Jiang, and Chang-Won Lee. "Heterosubtypic protection against avian influenza virus by live attenuated and chimeric norovirus P-particle-M2e vaccines in chickens." Vaccine 37, no. 10 (February 2019): 1356–64. http://dx.doi.org/10.1016/j.vaccine.2019.01.037.

Full text
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