Announcement

Collapse
No announcement yet.

PB1-F2 protein of Influenza A virus: increasing pathogenicity disrupting macrophages

Collapse
X
 
  • Filter
  • Time
  • Show
Clear All
new posts

  • PB1-F2 protein of Influenza A virus: increasing pathogenicity disrupting macrophages

    With the prospect of another pandemic Influenza fresh in our consciousness, the pathogenic nature of the Influenza A virus and its ability to induce high rates of mortality are ever more pertinent. Recently a novel protein encoded by an alternate reading frame in the PB1 Gene segment of Influenza A virus has been discovered and in turn shown to enhance viral virulence in a mouse model [1]. This protein has been shown to specifically target and destroy alveolar macrophages [2]. This review suggests that this protein, present in all previous pandemic strains, may reappear as a virulence factor in a subsequent pandemic strain. This PB1-F2 protein will enhance the mortality rate of the virus by increasing the likelihood of a secondary bacterial infection, which is the primary cause of death to a patient infected with Influenza A.


    Commentary

    The PB1-F2 protein of Influenza A virus: increasing pathogenicity by disrupting alveolar macrophages
    J Robert Coleman
    Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY 11794, USA

    Virology Journal 2007, 4:9 doi:10.1186/1743-422X-4-9

    The electronic version of this article is the complete one and can be found online at: http://www.virologyj.com/content/4/1/9

    <table class="smalltext"> <tbody> <tr> <td>Received</td> <td width="25"> </td> <td>8 January 2007</td></tr> <tr> <td>Accepted</td> <td width="25"> </td> <td>15 January 2007</td></tr> <tr> <td>Published</td> <td width="25"> </td> <td>15 January 2007</td></tr></tbody></table>
    ? 2007 Coleman; licensee BioMed Central Ltd.
    This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

    Abstract

    With the prospect of another pandemic Influenza fresh in our consciousness, the pathogenic nature of the Influenza A virus and its ability to induce high rates of mortality are ever more pertinent. Recently a novel protein encoded by an alternate reading frame in the PB1 Gene segment of Influenza A virus has been discovered and in turn shown to enhance viral virulence in a mouse model [1]. This protein has been shown to specifically target and destroy alveolar macrophages [2]. This review suggests that this protein, present in all previous pandemic strains, may reappear as a virulence factor in a subsequent pandemic strain. This PB1-F2 protein will enhance the mortality rate of the virus by increasing the likelihood of a secondary bacterial infection, which is the primary cause of death to a patient infected with Influenza A.

    Background


    An intrinsic property of all viruses is their need to either subvert and/or subdue the host immune response to establish a productive replication cycle. The mechanism by which this avoidance proceeds is greatly varied throughout the phyla of viruses; however either must occur in order for a virus to spread and propagate. The enhancement of this ability to subvert and disable host immunity is directly correlated to increased viral pathogenicity in the host [3]. For example the 3C viral protease of Poliovirus cleaves the p65-RelA subunit of NF-κB, a transcription factor involved in the cellular innate immune response [4]. Another human pathogen, Influenza A virus, has many well-elucidated mechanisms used to avoid both the innate and adaptive human immune responses. Since the current concern over the possible emergence of an Influenza A virus pandemic has arisen, Influenza A virus's mechanisms of evasion are of great significance. The main culprit of evasion on the cellular level is the NS1 protein, which is responsible for inactivation of the host innate immune response by preventing activation of PKR from INF-α/β signaling, thus allowing replication and viral protein synthesis to proceed unabated in the host cell [5]. Additionally, Influenza A virus has the ability to escape the host's humoral immunity by a phenomenon known as antigenic drift. This mutation driven phenomena produces changes both in amino acids and glycosylation patterns of the two virus envelope glycoproteins, hemagglutinin (HA) and neuraminidase (NA) [6]. This concept of antigenic drift is significant because it contributes to the continued, seasonal pathogenicity associated with epidemic Influenza A virus, i.e. the need for a new flu vaccine each year. Since Influenza manipulates and creates new antigenic determinates via mutations, the human population becomes steadily immunologically inept, where by circulating antibodies are incapable of, or have a reduced capacity to, neutralize a repeat infection. A more threatening avoidance of humoral and innate cellular immunity is the ability of Influenza A virus to reassort via the phenomena of antigenic shift. Since Influenza A virus is an eight-segmented minus-sense RNA virus, its segmented nature allows for the swapping and exchange of gene segments between different strains. Specifically this occurs by human influenza viruses swapping the HA glycoprotein, NA glycoprotein or polymerase (PB1, PB2, PA) segments with those of avian and pig Influenza A viruses. Therefore, a reassortment of gene segments has occurred creating an entirely novel Influenza A virus strain capable of infecting humans. [6]. In turn, the global human population is entirely immunologically na?ve to these novel viruses and these viruses become the cause of pandemics that result in a vast number of human deaths; the most notably of which was the 1918 Spanish Flu that killed an estimated 30 to 50 million people [7].


    Influenza A virus and the PB1-F2 protein


    Other additional mechanisms of host defense manipulation and avoidance by Influenza A virus exist including a novel alternate reading frame recently discovered in the PB1 polymerase gene segment. This reading frame is found in select Influenza A viruses and has been shown to impact host defense mechanisms and in turn enhance pathogenicity in vivo [1]. This protein, named PB1-F2, has an apoptotic induction effect on macrophages, thus reducing their ability to contribute to an immune response [8]. It has been previously suggested that this PB1-F2 protein contributes to viral pathogenicity solely because of it causes an inhibition of viral clearance, thus increasing cytoxicity [1]. However, it is possible that by directly targeting professional antigen presenting cells for destruction, the PB1-F2 can also contribute to Influenza A virus pathogenicity by increasing the probability of an opportunistic secondary bacterial infection. The major source of mortality associated with Influenza A infection is indeed these secondary bacterial infections [9].
    The discovery of a novel protein that contributes to an increase in Influenza A virus pathogenicity is of great concern due to the impact that Influenza A virus has on both human health as well as on the economy. Globally it is estimated that between 300,000 to 500,000 people may die annually due to influenza virus infections [10]. In the United States, influenza viruses are the cause of widespread mortality and morbidity. For example, influenza viruses cause approximately 35,000 deaths each year in the United States [11]. In addition to the impact on human health, influenza virus infections are responsible for an estimated 200,000 hospitalizations, thus impacting the United States economy by an approximate cost of 23 billion dollars each year [12]. Although a productive viral infection can elicit disease in all age groups, serious, life-threatening complications are markedly increased in children and elderly persons 65 years of age and over. By further elucidation of the various mechanisms that contribute to the pathogenic nature of Influenza A virus, there can be a beneficial impact on human health and the economy by way of enhancing vaccine development or increasing the effectiveness of antiviral drugs.
    Specifically, Influenza A virus is an enveloped, eight segmented minus-sense RNA virus, that is a member of the orthomyxoviridae genus of viruses. Unique to Influenza A virus, compared to other lytic mammalian RNA viruses, is that all mRNA synthesis and genome replication occurs in the nucleus of the infected host cell [13]. Furthermore, Influenza A virus replication proceeds rapidly, evident by the completion of a replication cycle in 10 hours. Primarily Influenza A virus infects the respiratory tract via the inhalation of aerosols containing infectious virions. The infection and replication of Influenza A virus is cytolytic and is usually limited to superficial epithelial cells of the upper and lower respiratory tract. However, secondary sites of infection can occur, such as infections of nervous tissue, which results in Reye's syndrome- a condition characterized by acute encephalopathy [14].


    PB1-F2 aiding in secondary bacterial infections?


    Despite Influenza A virus's speedy replication cycle and cytolytic properties, the major cause of death associated with an Influenza A virus infection is secondary bacterial pneumonia. The major pathogens associated with this complication are Streptococcus pneumoniae, Staphylococcus aureus, and Haemophilus influenzae [9]. One of the symptoms usually associated with flu, dry cough, is indicative of the loss of ciliated, mucous-producing epithelial cells as a result of the viral infection destroying these cells lining the respiratory tract [15]. The mucus produced by these cells in a normal, healthy individual serves to clear invading microorganisms as well as particulate matter [16]. However, during an Influenza A virus infection this clearance function is significantly impaired and therefore opportunistic bacteria can reside in the lung longer, and in turn establish a secondary infection which results in the increased death rate.
    Besides the destruction of mucus producing lung epithelia cells, another possible contribution to secondary bacterial infection as the result of Influenza A infection is the targeted elimination of alveolar macrophages by the Influenza PB1-F2 protein. Initially precursors to macrophages, monoblasts, are produced in the bone marrow and secreted into the bloodstream where they become monocytes. Once monocytes exit the circulation and migrate into tissue, they differentiate into tissue specific resident macrophages, such as Kuppfer cells in the liver or alveolar macrophages in the lung [17]. These resident macrophages serve an immuno-surveillance function as a part of innate immunity, constantly eliminating foreign antigens from tissues and organs [18]. Alveolar macrophages, which are located at the air-tissue interface in the lung, are the initial cells that interact with inhaled microorganisms and particulate matter [19]. During normal respiration a human breathes in an estimated 7,000 liters of air per day [20]. Therefore, the alveolar macrophage's rapid recognition of invading pathogens via pattern recognition receptors (PRR) recognizing pathogen associated molecular patterns (PAMPS) and subsequent phagocytosis, is vital to maintaining healthy tissue [21]. Any impairment of these cells by PB1-F2 would greatly facilitate the establishment of a bacterial pulmonary infection. Also these resident macrophages communicate with the adaptive arm of the immune response via antigen presentation to CD4<sup>+ </sup>T<sub>H </sub>cells. This antigen presentation results in clonal expansion of a given CD4<sup>+ </sup>T<sub>H </sub>cell lineage and subsequently a range of downstream effector functions are induced, such as cytotoxic T lymphocyte (CTL) activation, antibody production, and inflammation [22]. By PB1-F2 specifically targeting macrophages for elimination, the induction of the acquired immune response is delayed and debilitated. This will allow for reduced clearance of Influenza A virus. A prolonged presence in the host will increase damage to host tissue and also produce more infectious virions that will enable increased transmission. More importantly, any delay or impairment in the immune response by PB1-F2 mediated destruction of professional antigen presenting cells could facilitate an opportunistic bacterial infection.
    The initial discovery of the PB1-F2 protein by Chen et al. occurred via a broad search for antigenic Influenza A viral peptides that are encoded by alternate reading frames. Chen et al. also examined if these novel polypeptides are presented by major histocompatibility complex I and in turn recognized by CD8<sup>+ </sup>CTL on the surface of infected cells. After scanning the Influenza A genome for this novel antigenic, CTL-activating peptide, it was found to correspond to a protein encoded by an alternate reading frame found within the PB1 gene segment [2]. A simple schematic representation of the PB1-F2 can be seen in Fig. 1 adapted from Lamb et al. 2001. It is believed this alternate reading frame initiates at a start site (+) 120 base pairs upstream from the PB1 gene reading frame and is expressed possibly due to ribosomal scanning [23]. Ribosomal scanning is the process by which the 40S subunit of the ribosome, after loading directly downstream of the 5' cap by the translation machinery, "scans" for an AUG triplet to initiate translation; however if the ribosome recognizes an AUG in an alternate reading frame, a novel polypeptide could be produced [24]. When analyzing the PB1-F2 protein, it was found to be rather short lived in the replication cycle, with its maximum expression occurring approximately 5 hours post-infection [2]. The protein was found to localize to the inner and outer membranes of mitochondria via a basic amphipathic helix in its C-terminus [25]. When localized to the mitochondria, PB1-F2 can induce a dramatic degradation of mitochondrial morphology, which results in the reduction of the membrane potential and the induction of apoptosis [26]. PB1-F2 possibly functions by creating nonspecific pores in lipid bilayer membranes [27]. Interestingly this apoptotic induction occurs in monocytes much more readily than in epithelial cells, which was confirmed experimentally in tissue culture. This apoptotic induction can occur both when the protein is expressed intracellularly or simply present extracellularly to the macrophages [23]. It has also been shown that this PB1-F2 protein is not required for replication due to the viability in tissue culture of knockout viruses lacking the PB1-F2 reading frame [1]; interestingly the PB1-F2 knockout strain did show a marked decrease, approximately 50%, in their ability to induce monocyte apoptosis.
    In sum, these in vitro findings strongly suggested that the presence of the PB1-F2 protein would have an increasing effect on viral pathogenicity in vivo. This hypothesis was confirmed by a recent study conducted by Zamarin et al., which examined PB1-F2's contribution to Influenza A virus pathogenesis in a mouse model. Indeed a virus expressing the PB1-F2 was found to have an increased virulence when compared to a knockout virus lacking the alternate reading frame. Also the presence of higher viral titers in the lungs of mice infected with a PB1-F2 expressing strain demonstrated a reduction in the animal's ability to clear the virus as well as mount an effective immune response [1]. Since PB1-F2 is found to have an apoptotic effect on professional antigen presenting cells, the PB1-F2 mediated killing of these cells could impede antigen presentation to the adaptive arm of the immune response, thus allowing for the increased pathogenicity of the virus.
    Interestingly this alternate reading frame is found in the pandemic strains of 1968, 1957, and the infamous 1918 strain [1]. Indeed these viruses have a markedly pathogenic phenotype evident by the staggering death rates associated with each pandemic strain. All previous conclusions suggest that the increased pathogenicity of viruses encoding this protein is solely due to the virus's ability to reduce its own antigen presentation as well as reduce viral clearance. However, it is known that the major cause of death associated with an Influenza A virus infection is the establishment of a secondary bacterial infection [9]. Granted the cytotoxic, primary Influenza A infection is vastly damaging and PB1-F2 contributes to viral virulence by inducing apoptosis in macrophages. This targeted elimination of macrophages increases virulence by reducing antigen presentation and preventing crosstalk between the innate and adaptive arms of the immune system. However, the pathogenic enhancement of the PB1-F2 expressing viruses may also be due to an infected host's increased susceptibility to a secondary bacterial infection. This is an intriguing possibility and one that has been overlooked by previous studies.


    Conclusion


    Currently it has been found after large-scale, exhaustive sequence analysis of avian Influenza A virus isolates that the PB1-F2 transcript is under the highest positive selective pressure for nonsynonymous substitutions [28]. This combined with the emerging threat of a possible human pandemic of Influenza A virus brings the enhancement of pathogenicity by PB1-F2 to the forefront. The World Health Organization's (WHO) policy now is to recommend the stockpiling of antibiotics to combat secondary bacterial infections associated with an outbreak of pandemic Influenza A virus [29]. If one could also develop an antiviral antagonist of the PB1-F2 protein (especially due to its ability to induce macrophage apoptosis when present extracellularly) the targeted destruction of professional antigen presenting cells could be inhibited. Therefore, the ability to clear virus and more importantly to fend off opportunistic bacterial infections would be maintained. The inhibition of PB1-F2 could prove to have a profound effect on human health because this could reduce the high rates of mortality associated with pandemic and epidemic Influenza A viruses carrying this alternate reading frame.


    Acknowledgements


    The author would like to thank Lisa M. Runco for her editorial advice. Also the author is supported in part by The American Foundation for Aging Research.

    References

    <table class="bodytext" border="0" cellpadding="0" cellspacing="0" width="100%"><tbody><tr> <td align="right" valign="top">1.</td> <td width="5"> </td> <td colspan="2" valign="top">Zamarin D, et al.: Influenza A virus PB1-F2 protein contributes to viral pathogenesis in mice.
    J Virol 2006, 80(16):7976-83. [PubMed Abstract] [Publisher Full Text] [PubMed Central Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1] [2] [3] [4] [5] [6]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">2.</td> <td width="5"> </td> <td colspan="2" valign="top">Chen W: A novel influenza A virus mitochondrial protein that induces cell death.
    Nat Med 2001, 7(12):1306-12. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1] [2] [3]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">3.</td> <td width="5"> </td> <td colspan="2" valign="top">Smith H: Mechanisms of virus pathogenicity.
    Bacteriol Rev 1972, 36(3):291-310. [PubMed Abstract][PubMed Central Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">4.</td> <td width="5"> </td> <td colspan="2" valign="top">Neznanov N, et al.: Proteolytic cleavage of the p65-RelA subunit of NF-kappaB during poliovirus infection.
    J Biol Chem 280(25):24153-8. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">2005 Jun 24; Epub 2005 Apr 21
    Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">5.</td> <td width="5"> </td> <td colspan="2" valign="top">Salvatore M, et al.: Effects of influenza A virus NS1 protein on protein expression: the NS1 protein enhances translation and is not required for shutoff of host protein synthesis.
    J Virol 2002, 76(3):1206-12. [PubMed Abstract] [Publisher Full Text] [PubMed Central Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">6.</td> <td width="5"> </td> <td colspan="2" valign="top">Steinhauer DA, et al.: Genetics of influenza virus.
    Annu Rev Genet 2002, 36:305-332. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1] [2]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">7.</td> <td width="5"> </td> <td colspan="2" valign="top">Russell CJ, et al.: The genesis of a pandemic influenza virus.
    Cell 2005, 123:368-371. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">8.</td> <td width="5"> </td> <td colspan="2" valign="top">Zamarin D, et al.: Influenza virus PB1-F2 protein induces cell death through mitochondrial ANT3 and VDAC1.
    PLoS Pathog 2005, 1(1):e4. [PubMed Abstract] [Publisher Full Text] [PubMed Central Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Epub 2005 Sep 30
    Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">9.</td> <td width="5"> </td> <td colspan="2" valign="top">Sethi S: Bacterial pneumonia. Managing a deadly complication of influenza in older adults with comorbid disease.
    Geriatrics 2002, 57(3):56-61. [PubMed Abstract]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1] [2] [3]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">10.</td> <td width="5"> </td> <td colspan="2" valign="top">Kamps BS, Hoffmann C, Preiser W, (Eds): Influenza Report
    [http://www.InfluenzaReport.com.] In Flying Publisher: Paris; 2006.
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">11.</td> <td width="5"> </td> <td colspan="2" valign="top">Thompson WW, et al.: Mortality associated with influenza and respiratory syncytial virus in the United States.
    JAMA 2003, 289:179-186. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">12.</td> <td width="5"> </td> <td colspan="2" valign="top">Cram P, et al.: Influenza: Cost of illness and considerations in the economic evaluation of new and emerging therapies.
    Pharmacoeconomics 2001, 19:223-230. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">13.</td> <td width="5"> </td> <td colspan="2" valign="top">Neuman G, et al.: Orthomyxovirus replication, transcription, and polyadenylation.
    Curr Top Microbiol Immunol 2004, 283:121-43. [PubMed Abstract]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">14.</td> <td width="5"> </td> <td colspan="2" valign="top">Belay ED: Reye's syndrome in the United States from 1981 through 1997.
    N Engl J Med 340(18):1377-82. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">1999 May 6;
    Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">15.</td> <td width="5"> </td> <td colspan="2" valign="top">Bergen GA, et al.: Influenza. More than mom and chicken soup.
    J Fla Med Assoc 1996, 83(1):19-22. [PubMed Abstract]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">16.</td> <td width="5"> </td> <td colspan="2" valign="top">Chilvers MA: Local mucociliary defence mechanisms.
    Paediatr Respir Rev 2000, 1(1):27-34. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">17.</td> <td width="5"> </td> <td colspan="2" valign="top">van Furth R: Origin and turnover of monocytes and macrophages.
    Curr Top Pathol 1989, 79:125-150. [PubMed Abstract]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">18.</td> <td width="5"> </td> <td colspan="2" valign="top">Lehnert BE: Pulmonary and thoracic macrophage subpopulations and clearance of particles from the lung.
    Environ Health Perspect 1992, 97:17-46. [PubMed Abstract][PubMed Central Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">19.</td> <td width="5"> </td> <td colspan="2" valign="top">Sibille Y, et al.: Macrophages and polymorphonuclear neutrophils in lung defense and injury.
    Am Rev Respir Dis 1990, 141:471-501. [PubMed Abstract]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">20.</td> <td width="5"> </td> <td colspan="2" valign="top">Martin TR: Recognition of bacterial endotoxin in the lungs.
    Am J Respir Cell Mol Biol 2000, 23:128-132. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">21.</td> <td width="5"> </td> <td colspan="2" valign="top">Medzhitov R, et al.: Decoding the Patterns of Self and Nonself by the Innate Immune System.
    Science 296(5566):298-300. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">12 April 2002
    Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">22.</td> <td width="5"> </td> <td colspan="2" valign="top">Goldsby R, et al.: Kuby Immunology Cp2. 4th edition. W.H. Freeman And Company, New York; 2001:27.
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">23.</td> <td width="5"> </td> <td colspan="2" valign="top">Lamb R, et al.: Death by influenza virus protein.
    Nat Med 2001, 7(12):1286-8. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1] [2]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">24.</td> <td width="5"> </td> <td colspan="2" valign="top">Jackson RJ: Alternative mechanisms of initiating translation of mammalian mRNAs.
    Biochem Soc Trans 2005, 33(Pt 6):1231-41. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">25.</td> <td width="5"> </td> <td colspan="2" valign="top">Gibbs JS: The influenza A virus PB1-F2 protein targets the inner mitochondrial membrane via a predicted basic amphipathic helix that disrupts mitochondrial function.
    J Virol 2003, 77(13):7214-24. [PubMed Abstract] [Publisher Full Text] [PubMed Central Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">26.</td> <td width="5"> </td> <td colspan="2" valign="top">Yamada H, et al.: Mitochondrial targeting sequence of the influenza A virus PB1-F2 protein and its function in mitochondria.
    FEBs Lett 578(3):331-6. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">2004 Dec 17
    Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">27.</td> <td width="5"> </td> <td colspan="2" valign="top">Chanturiya N: PB1-F2, an influenza A virus-encoded proapoptotic mitochondrial protein, creates variably sized pores in planar lipid membranes.
    J Virol 2004, 78(12):6304-12. [PubMed Abstract] [Publisher Full Text] [PubMed Central Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">28.</td> <td width="5"> </td> <td colspan="2" valign="top">Obenauer JC, et al.: Large-scale sequence analysis of avian influenza isolates.
    311(5767):1576-80. [PubMed Abstract] [Publisher Full Text]
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">2006 Mar 17 Epub 2006 Jan 26
    Return to citation in text: [1]</td></tr> <tr> <td colspan="3"> </td></tr> <tr> <td align="right" valign="top">29.</td> <td width="5"> </td> <td colspan="2" valign="top">World Health Organization, Epidemic and Pandemic Alert and Response (EPR)
    Avian Influenza
    </td></tr> <tr> <td align="right" valign="top"> </td> <td width="5"> </td> <td colspan="2" valign="top">5 December 2005
    Return to citation in text: [1]</td></tr></tbody></table>

  • #2
    Re: PB1-F2 protein of Influenza A virus: increasing pathogenicity disrupting macropha

    <table width="1000"> <tbody> <tr> <td colspan="2" align="right" nowrap="nowrap">ColemanVirology Journal 2007 4:9 doi:10.1186/1743-422X-4-9</td></tr> <tr> <td align="left" nowrap="nowrap">Figure 1</td></tr></tbody></table>Click image for larger version

Name:	1743-422X-4-9-1-l.jpg
Views:	1
Size:	14.1 KB
ID:	648236

    The bisistronic PB1 gene segment of Influenza A Virus
    . Here we see the two open reading frames of the PB1 gene segment of Influenza A Virus. The red segment corresponds to the alternate reading frame that encodes the PB1-F2 protein whose start site is 120 bp downstream of the PB1 polymerase gene. This figure was adapted form Lamb et al. 2001 [23].

    Comment


    • #3
      Re: PB1-F2 protein of Influenza A virus: increasing pathogenicity disrupting macropha

      An interesting extract.

      Recently a novel protein encoded by an alternate reading frame in the PB1 Gene segment of Influenza A virus has been discovered and in turn shown to enhance viral virulence in a mouse model [1]. This protein has been shown to specifically target and destroy alveolar macrophages [2]. This review suggests that this protein, present in all previous pandemic strains, may reappear as a virulence factor in a subsequent pandemic strain. This PB1-F2 protein will enhance the mortality rate of the virus by increasing the likelihood of a secondary bacterial infection

      Comment


      • #4
        Re: PB1-F2 protein of Influenza A virus: increasing pathogenicity disrupting macropha

        Note also:
        Currently it has been found after large-scale, exhaustive sequence analysis of avian Influenza A virus isolates that the PB1-F2 transcript is under the highest positive selective pressure for nonsynonymous substitutions [28]. This combined with the emerging threat of a possible human pandemic of Influenza A virus brings the enhancement of pathogenicity by PB1-F2 to the forefront. The World Health Organization's (WHO) policy now is to recommend the stockpiling of antibiotics to combat secondary bacterial infections associated with an outbreak of pandemic Influenza A virus [29].
        The inhibition of PB1-F2 could prove to have a profound effect on human health because this could reduce the high rates of mortality associated with pandemic and epidemic Influenza A viruses carrying this alternate reading frame.

        Comment


        • #5
          Re: PB1-F2 protein of Influenza A virus: increasing pathogenicity disrupting macropha

          we had a thread here:
          I'm interested in expert panflu damage estimates
          my current links: http://bit.ly/hFI7H ILI-charts: http://bit.ly/CcRgT

          Comment


          • #6
            Re: PB1-F2 protein of Influenza A virus: increasing pathogenicity disrupting macropha

            Influenza A virus PB1-F2 gene


            Emerging Infectious Diseases, Oct, 2006 by Roland Zell, Andi Krumbholz, Peter Wutzler, Guang-Wu Chen, Shin-Ru Shih

            <!-- google_ad_section_start -->
            <!-- if there is a query term, show widget --> <!-- code re-engineered from partners/us/search_art_main --> Find More Results for: "PB1-F2 "
            Influenza A virus...
            Newfound flu protein...
            Cause of deadlier flus...




            <!-- no query term? print empty widget --> To the Editor: Recently, Chen and co-workers described the expression of an 11th influenza A virus protein, designated PB1-F2 because this protein is encoded in the +1 open reading frame of the segment-2 RNA (1). Later, Chen et al. presented a preliminary analysis of 336 PB1 sequences from GenBank (2). We have extended the work on PB1-F2 and analyzed 1,864 partial and complete segment-2 sequences deposited in GenBank; these sequences belong to 79 influenza A virus subtypes. In summary, the following 8 observations should receive attention:
            First, the size of PB1-F2 polypeptides ranges from 79 to 101 amino acids (aa); most isolates encode versions of either 87 or 90 aa. Because polypeptides of 79 aa are located within mitochondria, their truncation has no effect on the protein function. The frequency of the 79-aa PB1-F2 is [approximately equal to] 5%.
            Second, a functional PB1-F2 is expressed by 92% of all segment-2 sequences, i.e., a polypeptide >78 aa. The proportion of intact PB1-F2 varies according to host (humans 90%, swine 76%, other mammals 100%, birds 95%).
            Advertisement

            Third, the H1N1 subtype comprises 3 genetic lineages. One clade has 2 branches: 1 branch includes the human viruses, with the pandemic 1918 virus at its root; the other branch includes the classic swine viruses. The third clade represents the European porcine isolates. Although all classic swine sequences have a truncated PB1-F2 (in-frame stop codons after 11, 24, and 35 codons), the early human isolates (H1N1 sequences from 1918 through 1947) have an intact PB1-F2. After 1956, however, a mutation became prevalent such that the recent sequences starting from A/Beijing/1/56 terminate after 57 codons. An exception to this rule is A/Taiwan/3355/97. Two human H1N1 isolates with an intact PB1-F2 coding sequence cluster in the H3N2 clade (A/Kiev/59/79, A/Wisconsin/10/98). The PB1 sequences of European porcine influenza A virus isolates cluster with European porcine H3N2 and H1N2.
            Fourth, all H2N2 sequences are monophyletic and encode an intact PB1-F2. Fifth, the main sequence cluster of the H3N2 subtype comprises 3 branches: 1) porcine H3N2 and porcine H1N2 sequences from the United States, 2) porcine H3N2 isolates from Hong Kong and human H1N2, and 3) recent human H3N2 and some Japanese H3N2 isolates. Most of these sequences encode an intact PB1-F2.
            Sixth, the cluster of European porcine influenza A virus isolates comprises the subtypes H1N1, H1N2, and H3N2. The lack of distinct clades for each subtype indicates frequent reassortment in the evolution of these viruses. Of the segment-2 sequences, 56% encode an intact PB1-F2.
            Seventh, other porcine isolates of various subtypes represent trans-species infections or single reassortment events. And eighth, the segment-2 sequences of many avian influenza A virus isolates encode intact PB1-F2. Considerable proportions of truncated PB1-F2 genes were found in the H5N2, H6N6, H9N2, and H13N2 subtypes. However, because of the small number of sequences available, this observation may not be important.
            In conclusion, PB1-F2 is expressed in most avian and many porcine influenza A virus isolates. This finding contrasts with those in the initial publication, which stated that PB1-F2 is not expressed in many animal isolates, particularly those of porcine origin (1). Because PB1-F2 was described as a proapoptotic protein probably counteracting the host immune response, why numerous human and porcine isolates lack this protein without selective disadvantage remains unclear.
            Roland Zell, * Andi Krumbholz, * and Peter Wutzler *
            * Friedrich Schiller University, Jena, Germany
            References
            (1.) Chen W, Calvo PA, Malide D, Gibbs J, Schubert U, Bacik I, et al. A novel influenza A virus mitochondrial protein that induces cell death. Nat Med. 2001;7:1306-12.
            (2.) Chen GW, Yang CC, Ysao KC, Huang CG, Lee LA, Yang WZ, et al. Influenza A virus PB1-F2 gene in recent Taiwanese isolates. Emerg Infect Dis. 2004;10:630-6.
            Address for correspondence: Roland Zell, Institute of Virology and Antiviral Therapy, Medical Centre at the Friedrich Schiller University, Hans Knoell Str 2, D-07745 Jena, Germany; email: Roland.Zell@med.uni-jena.de
            In response: Zell et al. (1) performed an extensive genetic investigation of PB1-F2, based on up-to-date GenBank sequences. Their sample size (1,864) greatly outnumbered ours (336) in a previous study (2) and thus definitely better portrays the genetic characteristics of PB1-F2. We appreciate their analyzing these samples by subdividing nonhuman strains into different species, which we did not do (2). Their analysis is especially meaningful for the global pandemic threat from avian influenza viruses, which increases the need to study interspecies adaptation and transmission.
            Zell et al. found that 92% of PB1 RNA encodes a functional PB1-F2, compared with our 79% (264/334), which supports the increasingly crucial role of PB1-F2 in influenza virology. They found the proportion of intact human PB1-F2 to be 90%, a substantial boost from our 68% (67/99), which was based on data from late 2003 (2). This increase is apparently caused by the increasing number of human H3N2 sequences (mostly encoding an intact PB1-F2 compared with H1N1) deposited in the past 2 years.
            __________________________________________________ ________


            <dl class="PubmedArticle" id="dlPubmedArticle16873254"><dt class="head" id="head16873254">: J Virol. 2006 Aug;80(16):7976-83. <script language="JavaScript1.2"> <!-- var PopUpMenu2_LocalConfig_jsmenu3Config = [ ["ShowCloseIcon","yes"], ["Help","window.open('/entrez/query/static/popup.html','Links_Help','resizable=no,scrollbars= yes,toolbar=no,location=no,directories=no,status=n o,menubar=no,copyhistory=no,alwaysRaised=no,depend =no,width=400,height=500');"], ["FrameTarget","_top"], ["TitleText"," Links "] ] var jsmenu3Config = [ ["UseLocalConfig","jsmenu3Config","",""] ] //--> </script> <script language="JavaScript1.2"> <!-- var Menu16873254 = [ ["UseLocalConfig","jsmenu3Config","",""], ["Gene","http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?itool=pubmed_AbstractPlus&db=pubmed&cmd =Display&dopt=pubmed_gene&from_uid=16873254","",""], ["Gene (GeneRIF)","http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?itool=pubmed_AbstractPlus&db=pubmed&cmd =Display&dopt=pubmed_gene_rif&from_uid=16873254"," ",""], ["Cited Articles","http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?itool=pubmed_AbstractPlus&db=pubmed&cmd =Display&dopt=pubmed_pubmed_refs&from_uid=16873254 ","",""], ["Taxonomy via GenBank","http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?itool=pubmed_AbstractPlus&db=pubmed&cmd =Display&dopt=pubmed_taxonomy_entrez&from_uid=1687 3254","",""], ["Free in PMC","http://www.pubmedcentral.gov/articlerender.fcgi?tool=pubmed&pubmedid=16873254", "",""], ["Cited in PMC","http://www.pubmedcentral.gov/tocrender.fcgi?action=cited&tool=pubmed&pubmedid=1 6873254","",""], ["LinkOut","http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?itool=pubmed_AbstractPlus&cmd=Retrieve& db=pubmed&list_uids=16873254&dopt=ExternalLink","" ,""] ] //--> </script> Links
            </dt><dd class="abstract" id="abstract16873254">Influenza A virus PB1-F2 protein contributes to viral pathogenesis in mice.
            Department of Microbiology, Box 1124, Mount Sinai School of Medicine, 1 Gustave Levy Place, New York, NY 10029, USA.
            The influenza virus PB1-F2 protein is a novel protein previously shown to be involved in induction of cell death. Here we characterize the expression and the function of the protein within the context of influenza viral infection in tissue culture and a mouse model. We show that the C-terminal region of the protein can be expressed from a downstream initiation codon and is capable of interaction with the full-length protein. Using this knowledge, we generated influenza viruses knocked out for the expression of PB1-F2 protein and its downstream truncation products. Knocking out the PB1-F2 protein had no effect on viral replication in tissue culture but diminished virus pathogenicity and mortality in mice. The viruses replicated to similar levels in mouse lungs by day 3 postinfection, suggesting that the knockout did not impair viral replication. However, while the PB1-F2 knockout viruses were cleared after day 5, the wild-type viruses were detectable in mouse lungs until day 7, implying that expression of PB1-F2 resulted in delayed clearance of the viruses by the host immune system. Based on our findings and on the fact that the PB1 genomic segment was always newly introduced into some pandemic influenza viruses of the last century, we speculate that the PB1-F2 protein plays an important role in pathogenesis of influenza virus infection and may be an important contributor to pathogenicity of pandemic influenza viruses.
            PMID: 16873254 [PubMed - indexed for MEDLINE]
            </dd></dl>

            Comment

            Working...
            X