Displaying publications 1 - 20 of 41 in total

Abstract:
Sort:
  1. Al-qattan MN, Mordi MN
    J Mol Model, 2010 May;16(5):975-91.
    PMID: 19856192 DOI: 10.1007/s00894-009-0606-y
    In this study fragment-based drug design is combined with molecular docking simulation technique, to design databases of virtual sialic acid (SA) analogues with new substitutions at C2, C5 and C6 positions of SA scaffold. Using spaces occupied by C2, C5 and C6 natural moieties of SA when bound to hemagglutinin (HA) crystallographic structure, new fragments that are commercially available were docked independently in all the pockets. The oriented fragments were then connected to the SA scaffold with or without incorporation of linker molecules. The completed analogues were docked to the whole SA binding site to estimate their binding conformations and affinities, generating three databases of HA-bound SA analogues. Selected new analogues showed higher estimated affinities than the natural SA when tested against H3N2, H5N1 and H1N1 subtypes of influenza A. An improvement in the binding energies indicates that fragment-based drug design when combined with molecular docking simulation is capable to produce virtual analogues that can become lead compound candidates for anti-flu drug discovery program.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/immunology; Influenza A Virus, H5N1 Subtype/metabolism*; Influenza A Virus, H5N1 Subtype/chemistry
  2. Alexander DJ
    Avian Dis, 2007 Mar;51(1 Suppl):161-6.
    PMID: 17494548
    Between December 2003 and January 2004 highly pathogenic avian influenza (HPAI) H5N1 infections of poultry were declared in China, Japan, South Korea, Laos, Thailand, Cambodia, Vietnam, and Indonesia. In 2004 an outbreak was reported in Malaysia. In 2005 H5N1 outbreaks were recorded in poultry in Russia, Kazakhstan, Mongolia, Romania, Turkey, and Ukraine, and virus was isolated from swans in Croatia. In 2004 HPAI H5N1 virus was isolated from smuggled eagles detected at the Brussels Airport and in 2005 imported caged birds held in quarantine in England. In 2006 HPAI was reported in poultry in Iraq, India, Azerbaijan, Pakistan, Myanmar, Afghanistan, and Israel in Asia; Albania, France, and Sweden in Europe; and Nigeria, Cameroon, and Niger in Africa; as well as in wild birds in some 24 countries across Asia and Europe. In 2003, over 25,000,000 birds were slaughtered because of 241 outbreaks of HPAI caused by virus of H7N7 subtype in the Netherlands. The virus spread into Belgium (eight outbreaks) and Germany (one outbreak). HPAI H5N2 virus was responsible for outbreaks in ostriches in South Africa during 2005. HPAI H7N3 virus was isolated in Pakistan in 2004. Low-pathogenicity avian influenza (LPAI) H5 or H7 viruses were isolated from poultry in Italy (H7N3 2002-2003; H5N2 2005), The Netherlands (H7N3 2002), France (H5N2 2003), Denmark (H5N7 2003), Taiwan (H5N2 2004), and Japan (H5N2 2005). Many isolations of LPAI viruses of other subtypes were reported from domestic and wild birds. Infections with H9N2 subtype viruses have been widespread across Asia during 2002-06.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/classification; Influenza A Virus, H5N1 Subtype/isolation & purification*; Influenza A Virus, H5N1 Subtype/pathogenicity
  3. Eagles D, Siregar ES, Dung DH, Weaver J, Wong F, Daniels P
    Rev. - Off. Int. Epizoot., 2009 Apr;28(1):341-8.
    PMID: 19618637
    Since the first H5N1 highly pathogenic avian influenza virus (HPAIV) infection in the region in August 2003, Cambodia, Laos, Malaysia, Myanmar, Indonesia, Thailand and Vietnam have all recorded outbreaks of the disease. The HPAIV continues to occur in some countries in Southeast Asia despite control programmes encompassing surveillance, vaccination and stamping out strategies. A number of strains have been circulating in the region since the first outbreaks in 2003, and although the source of the initial outbreaks in domestic poultry is not known, the continuing propagation of disease in the region is primarily the result of the movement of domestic poultry and poultry products, and people. A comprehensive approach using all the strategies available to break the chain of transmission of the virus in poultry will be needed to achieve lasting disease control.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/classification; Influenza A Virus, H5N1 Subtype/pathogenicity*
  4. Ng LF, Barr I, Nguyen T, Noor SM, Tan RS, Agathe LV, et al.
    BMC Infect Dis, 2006;6:40.
    PMID: 16512903
    Continuous outbreaks of the highly pathogenic H5N1 avian influenza A in Asia has resulted in an urgent effort to improve current diagnostics to aid containment of the virus and lower the threat of a influenza pandemic. We report here the development of a PCR-based assay that is highly specific for the H5N1 avian influenza A virus.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/genetics*; Influenza A Virus, H5N1 Subtype/isolation & purification*
  5. Tan TS, Syed Hassan S, Yap WB
    Lett Appl Microbiol, 2017 Jun;64(6):446-451.
    PMID: 28370088 DOI: 10.1111/lam.12738
    The study aimed to construct a recombinant Lactobacillus casei expressing the nonstructural (NS) 1 protein of influenza A virus H5N1 on its cell wall. The NS1 gene was first amplified and fused to the pSGANC332 expression plasmid. The NS1 protein expression was carried out by Lact. casei strain C1. PCR screening and DNA sequencing confirmed the presence of recombinant pSG-NS1-ANC332 plasmid in Lact. casei. The plasmid was stably maintained (98·94 ± 1·65%) by the bacterium within the first 20 generations without selective pressure. The NS1 was expressed as a 49-kDa protein in association with the anchoring peptide. The yield was 1·325 ± 0·065 μg mg(-1) of bacterial cells. Lactobacillus casei expressing the NS1 on its cell wall was red-fluorescently stained, but the staining was not observed on Lact. casei carrying the empty pSGANC332. The results implied that Lact. casei strain C1 is a promising host for the expression of surface-bound NS1 protein using the pSGANC332 expression plasmid.

    SIGNIFICANCE AND IMPACT OF THE STUDY: The study has demonstrated, for the first time, the expression of nonstructural 1 (NS1) protein of influenza A virus H5N1 on the cell wall of Lactobacillus casei using the pSGANC332 expression plasmid. Display of NS1 protein on the bacterial cell wall was evident under an immunofluorescence microscopic observation. Lactobacillus casei carrying the NS1 protein could be developed into a universal oral influenza vaccine since the NS1 is highly conserved among influenza viruses.

    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/genetics; Influenza A Virus, H5N1 Subtype/immunology*
  6. Wang J, Vijaykrishna D, Duan L, Bahl J, Zhang JX, Webster RG, et al.
    J Virol, 2008 Apr;82(7):3405-14.
    PMID: 18216109 DOI: 10.1128/JVI.02468-07
    The transmission of highly pathogenic avian influenza H5N1 virus to Southeast Asian countries triggered the first major outbreak and transmission wave in late 2003, accelerating the pandemic threat to the world. Due to the lack of influenza surveillance prior to these outbreaks, the genetic diversity and the transmission pathways of H5N1 viruses from this period remain undefined. To determine the possible source of the wave 1 H5N1 viruses, we recently conducted further sequencing and analysis of samples collected in live-poultry markets from Guangdong, Hunan, and Yunnan in southern China from 2001 to 2004. Phylogenetic analysis of the hemagglutinin and neuraminidase genes of 73 H5N1 isolates from this period revealed a greater genetic diversity in southern China than previously reported. Moreover, results show that eight viruses isolated from Yunnan in 2002 and 2003 were most closely related to the clade 1 virus sublineage from Vietnam, Thailand, and Malaysia, while two viruses from Hunan in 2002 and 2003 were most closely related to viruses from Indonesia (clade 2.1). Further phylogenetic analyses of the six internal genes showed that all 10 of those viruses maintained similar phylogenetic relationships as the surface genes. The 10 progenitor viruses were genotype Z and shared high similarity (>/=99%) with their corresponding descendant viruses in most gene segments. These results suggest a direct transmission link for H5N1 viruses between Yunnan and Vietnam and also between Hunan and Indonesia during 2002 and 2003. Poultry trade may be responsible for virus introduction to Vietnam, while the transmission route from Hunan to Indonesia remains unclear.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/classification*; Influenza A Virus, H5N1 Subtype/genetics*; Influenza A Virus, H5N1 Subtype/isolation & purification
  7. Hurt AC, Selleck P, Komadina N, Shaw R, Brown L, Barr IG
    Antiviral Res, 2007 Mar;73(3):228-31.
    PMID: 17112602
    Since 2003, highly pathogenic A(H5N1) influenza viruses have been the cause of large-scale death in poultry and the subsequent infection and death of over 140 humans. A group of 55 influenza A(H5N1) viruses isolated from various regions of South East Asia between 2004 and 2006 were tested for their susceptibility to the anti-influenza drugs the neuraminidase inhibitors and adamantanes. The majority of strains were found to be fully sensitive to the neuraminidase inhibitors oseltamivir carboxylate, zanamivir and peramivir; however two strains demonstrated increased IC50 values. Sequence analysis of these strains revealed mutations in the normally highly conserved residues 116 and 117 of the N1 neuraminidase. Sequence analysis of the M2 gene showed that all of the A(H5N1) viruses from Vietnam, Malaysia and Cambodia contained mutations (L26I and S31N) associated with resistance to the adamantane drugs (rimantadine and amantadine), while strains from Indonesia were found to be a mix of both adamantane resistant (S31N) and sensitive viruses. None of the A(H5N1) viruses from Myanmar contained mutations known to confer adamantane resistance. These results support the use of neuraminidase inhibitors as the most appropriate class of antiviral drug to prevent or treat human A(H5N1) virus infections.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/drug effects*; Influenza A Virus, H5N1 Subtype/genetics; Influenza A Virus, H5N1 Subtype/isolation & purification
  8. Balasubramaniam VR, Wai TH, Omar AR, Othman I, Hassan SS
    Virol J, 2012;9:53.
    PMID: 22361110 DOI: 10.1186/1743-422X-9-53
    Highly-pathogenic avian influenza (HPAI) H5N1 and Newcastle disease (ND) viruses are the two most important poultry viruses in the world, with the ability to cause classic central nervous system dysfunction in poultry and migratory birds. To elucidate the mechanisms of neurovirulence caused by these viruses, a preliminary study was design to analyze host's cellular responses during infections of these viruses.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/pathogenicity*
  9. Loh LC, Hui DS, Beasley R
    Respirology, 2008 Mar;13 Suppl 1:S1.
    PMID: 18366520 DOI: 10.1111/j.1440-1843.2008.01245.x
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype*
  10. Wkly. Epidemiol. Rec., 2006 Feb 24;81(8):69-70.
    PMID: 16671220
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype*
  11. Hasan NH, Ebrahimie E, Ignjatovic J, Tarigan S, Peaston A, Hemmatzadeh F
    PLoS One, 2016;11(6):e0156418.
    PMID: 27362795 DOI: 10.1371/journal.pone.0156418
    A common approach for developing diagnostic tests for influenza virus detection is the use of mouse or rabbit monoclonal and/or polyclonal antibodies against a target antigen of the virus. However, comparative mapping of the target antigen using antibodies from different animal sources has not been evaluated before. This is important because identification of antigenic determinants of the target antigen in different species plays a central role to ensure the efficiency of a diagnostic test, such as competitive ELISA or immunohistochemistry-based tests. Interest in the matrix 2 ectodomain (M2e) protein of avian influenza virus (AIV) as a candidate for a universal vaccine and also as a marker for detection of virus infection in vaccinated animals (DIVA) is the rationale for the selection of this protein for comparative mapping evaluation. This study aimed to map the epitopes of the M2e protein of avian influenza virus H5N1 using chicken, mouse and rabbit monoclonal or monospecific antibodies. Our findings revealed that rabbit antibodies (rAbs) recognized epitope 6EVETPTRN13 of the M2e, located at the N-terminal of the protein, while mouse (mAb) and chicken antibodies (cAbs) recognized epitope 10PTRNEWECK18, located at the centre region of the protein. The findings highlighted the difference between the M2e antigenic determinants recognized by different species that emphasized the importance of comparative mapping of antibody reactivity from different animals to the same antigen, especially in the case of multi-host infectious agents such as influenza. The findings are of importance for antigenic mapping, as well as diagnostic test and vaccine development.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/immunology*
  12. Smith GJ, Fan XH, Wang J, Li KS, Qin K, Zhang JX, et al.
    Proc Natl Acad Sci U S A, 2006 Nov 07;103(45):16936-41.
    PMID: 17075062
    The development of highly pathogenic avian H5N1 influenza viruses in poultry in Eurasia accompanied with the increase in human infection in 2006 suggests that the virus has not been effectively contained and that the pandemic threat persists. Updated virological and epidemiological findings from our market surveillance in southern China demonstrate that H5N1 influenza viruses continued to be panzootic in different types of poultry. Genetic and antigenic analyses revealed the emergence and predominance of a previously uncharacterized H5N1 virus sublineage (Fujian-like) in poultry since late 2005. Viruses from this sublineage gradually replaced those multiple regional distinct sublineages and caused recent human infection in China. These viruses have already transmitted to Hong Kong, Laos, Malaysia, and Thailand, resulting in a new transmission and outbreak wave in Southeast Asia. Serological studies suggest that H5N1 seroconversion in market poultry is low and that vaccination may have facilitated the selection of the Fujian-like sublineage. The predominance of this virus over a large geographical region within a short period directly challenges current disease control measures.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/genetics*; Influenza A Virus, H5N1 Subtype/immunology; Influenza A Virus, H5N1 Subtype/isolation & purification; Influenza A Virus, H5N1 Subtype/pathogenicity
  13. Yusuf M, Mohamed N, Mohamad S, Janezic D, Damodaran KV, Wahab HA
    J Chem Inf Model, 2016 Jan 25;56(1):82-100.
    PMID: 26703840 DOI: 10.1021/acs.jcim.5b00331
    Increased reports of oseltamivir (OTV)-resistant strains of the influenza virus, such as the H274Y mutation on its neuraminidase (NA), have created some cause for concern. Many studies have been conducted in the attempt to uncover the mechanism of OTV resistance in H274Y NA. However, most of the reported studies on H274Y focused only on the drug-bound system, so the direct effects of the mutation on NA itself prior to drug binding still remain unclear. Therefore, molecular dynamics simulations of NA in apo form, followed by principal component analysis and interaction energy calculations, were performed to investigate the structural changes of the NA binding site as a result of the H274Y mutation. It was observed that the disruption of the NA binding site due to the H274Y mutation was initiated by the repulsive effect of Y274 on the 250-loop, which in turn altered the hydrogen-bonding network around residue 274. The rotated W295 side chain caused the upward movement of the 340-loop. Consequently, sliding box docking results suggested that the binding pathway of OTV was compromised because of the disruption of this binding site. This study also highlighted the importance of the functional group at C6 of the sialic acid mimicry. It is hoped that these results will improve the understanding of OTV resistance and shed some light on the design of a novel anti-influenza drug.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/drug effects; Influenza A Virus, H5N1 Subtype/enzymology
  14. Balasubramaniam VR, Hassan SS, Omar AR, Mohamed M, Noor SM, Mohamed R, et al.
    Virol J, 2011;8:196.
    PMID: 21529348 DOI: 10.1186/1743-422X-8-196
    Highly pathogenic Avian Influenza (HPAI) virus is able to infect many hosts and the virus replicates in high levels in the respiratory tract inducing severe lung lesions. The pathogenesis of the disease is actually the outcome of the infection as determined by complex host-virus interactions involving the functional kinetics of large numbers of participating genes. Understanding the genes and proteins involved in host cellular responses are therefore, critical for the elucidation of the mechanisms of infection.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/growth & development; Influenza A Virus, H5N1 Subtype/pathogenicity*
  15. Chaharaein B, Omar AR, Aini I, Yusoff K, Hassan SS
    Microbiol Res, 2009;164(2):174-9.
    PMID: 17336046
    Subtype-specific multiplex reverse transcription-polymerase chain reaction (RT-PCR) was developed to simultaneously detect three subtypes (H5, H7 and H9) of avian influenza virus (AIV) type A. The sensitivity of the multiplex RT-PCR was evaluated and compared to that of RT-PCR-enzyme-linked immunosorbent assay (ELISA) and conventional RT-PCR. While the sensitivity of the multiplex RT-PCR is as sensitive as the conventional RT-PCR, it is 10 times less sensitive than RT-PCR-ELISA. The multiplex RT-PCR is also as sensitive as the virus isolation method in detecting H9N2 from tracheal samples collected at day 3 and 5 post inoculation. Hence, the developed multiplex RT-PCR assay is a rapid, sensitive and specific assay for detecting of AIV subtypes.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/genetics; Influenza A Virus, H5N1 Subtype/isolation & purification
  16. Sims LD
    Avian Dis, 2007 Mar;51(1 Suppl):174-81.
    PMID: 17494550
    Numerous lessons have been learned so far in controlling H5N1 avian influenza in Asia. Early detection of incursions of virus prevented establishment of the disease in several countries, notably Japan, South Korea, and Malaysia. In countries where detection of early cases was delayed, infection is endemic and has been for three or more years. Control measures implemented in these countries need to reflect this finding. Vaccination will continue to be one of the key measures used in these endemically infected countries. Used alone, vaccination will not result in elimination of H5N1 viruses from a country, but, if used correctly, it will markedly reduce the prevalence of and susceptibility to infection. Vaccination has already played a valuable role in reducing the adverse effects of H5N1 viruses. Mass culling also reduces the level of infection in infected areas. However, the long-term benefits are limited in endemically infected countries owing to the high probability of reinfection on restocking unless other measures are used in parallel. Full epidemiological studies have not been conducted in many infected countries. Nevertheless, it is recognized that the number of clinical cases does not truly reflect the levels of infection. Domestic ducks and large live poultry markets have played a key role in the persistence of infection, because they can be infected silently. In tackling this disease, countries should adopt integrated control programs using the combination of measures best suited to the local environment. All surveillance data should be shared, both positive and negative, and should include information on cases of infection and disease. Socioeconomic and ecological implications of all control measures should be assessed before implementation, especially the impact on the rural poor.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/isolation & purification; Influenza A Virus, H5N1 Subtype/physiology*
  17. Chee Wei T, Nurul Wahida AG, Shaharum S
    Trop Biomed, 2014 Dec;31(4):792-801.
    PMID: 25776606 MyJurnal
    Malaysia first reported H5N1 poultry case in 2004 and subsequently outbreak in poultry population in 2007. Here, a recombinant gene encoding of peptide epitopes, consisting fragments of HA1, HA2 and a polybasic cleavage site of H5N1 strain Malaysia, was amplified and cloned into pET-47b(+) bacterial expression vector. DNA sequencing and alignment analysis confirmed that the gene had no alteration and in-frame to the vector. Then, His-tagged truncated HA protein was expressed in Escherichia coli BL21 (DE3) under 1 mM IPTG induction. The protein expression was optimized under a time-course induction study and further purified using Ni-NTA agarose under reducing condition. Migration size of protein was detected at 15 kDa by Western blot using anti-His tag monoclonal antibody and demonstrated no discrepancy compared to its calculated molecular weight.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/genetics*; Influenza A Virus, H5N1 Subtype/isolation & purification
  18. Jazayeri SD, Ideris A, Zakaria Z, Omar AR
    J Biomed Biotechnol, 2012;2012:264986.
    PMID: 22701301 DOI: 10.1155/2012/264986
    Attenuated Salmonella has been used as a carrier for DNA vaccine. However, in vitro and in vivo studies on the bacteria following transfection of plasmid DNA were poorly studied. In this paper, eukaryotic expression plasmids encoding avian influenza virus (AIV) subtype H5N1 genes, pcDNA3.1/HA, NA, and NP, were transfected into an attenuated Salmonella enteric typhimurium SV4089. In vitro stability of the transfected plasmids into Salmonella were over 90% after 100 generations. The attenuated Salmonella were able to invade MCF-7 (1.2%) and MCF-10A (0.5%) human breast cancer cells. Newly hatched specific-pathogen-free (SPF) chicks were inoculated once by oral gavage with 10(9) colony-forming unit (CFU) of the attenuated Salmonella. No abnormal clinical signs or deaths were recorded after inoculation. Viable bacteria were detected 3 days after inoculation by plating from spleen, liver, and cecum. Fluorescent in situ hybridization (FISH) and polymerase chain reaction (PCR) were carried out for confirmation. Salmonella was not detected in blood cultures although serum antibody immune responses to Salmonella O antiserum group D1 factor 1, 9, and 12 antigens were observed in all the inoculated chickens after 7 days up to 35 days. Our results showed that live attenuated S. typhimurium SV4089 harboring pcDNA3.1/HA, NA, and NP may provide a unique alternative as a carrier for DNA oral vaccine in chickens.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/genetics; Influenza A Virus, H5N1 Subtype/immunology
  19. Abubakar MB, Aini I, Omar AR, Hair-Bejo M
    J Biomed Biotechnol, 2011;2011:414198.
    PMID: 21541235 DOI: 10.1155/2011/414198
    Avian influenza (AI) is a highly contagious and rapidly evolving pathogen of major concern to the poultry industry and human health. Rapid and accurate detection of avian influenza virus is a necessary tool for control of outbreaks and surveillance. The AI virus A/Chicken/Malaysia/5858/2004 (H5N1) was used as a template to produce DNA clones of the full-length NS1 genes via reverse transcriptase synthesis of cDNA by PCR amplification of the NS1 region. Products were cloned into pCR2.0 TOPO TA plasmid and subsequently subcloned into pPICZαA vector to construct a recombinant plasmid. Recombinant plasmid designated as pPICZαA-NS1 gene was confirmed by PCR colony screening, restriction enzyme digestion, and nucleotide sequence analysis. The recombinant plasmid was transformed into Pichia pastoris GS115 strain by electroporation, and expressed protein was identified by SDS-PAGE and western blotting. A recombinant protein of approximately ~28 kDa was produced. The expressed protein was able to bind a rabbit polyclonal antibody of nonstructural protein (NS1) avian influenza virus H5N1. The result of the western blotting and solid-phase ELISA assay using H5N1 antibody indicated that the recombinant protein produced retained its antigenicity. This further indicates that Pichia pastoris could be an efficient expression system for a avian influenza virus nonstructural (NS1).
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/genetics*; Influenza A Virus, H5N1 Subtype/pathogenicity*
  20. Yusuf M, Konc J, Sy Bing C, Trykowska Konc J, Ahmad Khairudin NB, Janezic D, et al.
    J Chem Inf Model, 2013 Sep 23;53(9):2423-36.
    PMID: 23980878 DOI: 10.1021/ci400421e
    ProBiS is a new method to identify the binding site of protein through local structural alignment against the nonredundant Protein Data Bank (PDB), which may result in unique findings compared to the energy-based, geometry-based, and sequence-based predictors. In this work, binding sites of Hemagglutinin (HA), which is an important target for drugs and vaccines in influenza treatment, have been revisited by ProBiS. For the first time, the identification of conserved binding sites by local structural alignment across all subtypes and strains of HA available in PDB is presented. ProBiS finds three distinctive conserved sites on HA's structure (named Site 1, Site 2, and Site 3). Compared to other predictors, ProBiS is the only one that accurately defines the receptor binding site (Site 1). Apart from that, Site 2, which is located slightly above the TBHQ binding site, is proposed as a potential novel conserved target for membrane fusion inhibitor. Lastly, Site 3, located around Helix A at the stem domain and recently targeted by cross-reactive antibodies, is predicted to be conserved in the latest H7N9 China 2013 strain as well. The further exploration of these three sites provides valuable insight in optimizing the influenza drug and vaccine development.
    Matched MeSH terms: Influenza A Virus, H5N1 Subtype/drug effects; Influenza A Virus, H5N1 Subtype/immunology; Influenza A Virus, H5N1 Subtype/physiology
Filters
Contact Us

Please provide feedback to Administrator (afdal@afpm.org.my)

External Links