Neuraminidase of Influenza A Virus Binds Lysosome-Associated Membrane Proteins Directly and Induces Lysosome Rupture

J Virol. 2015 Oct;89(20):10347-58. doi: 10.1128/JVI.01411-15. Epub 2015 Aug 5.

Abstract

As a recycling center, lysosomes are filled with numerous acid hydrolase enzymes that break down waste materials and invading pathogens. Recently, lysosomal cell death has been defined as "lysosomal membrane permeabilization and the consequent leakage of lysosome contents into cytosol." Here, we show that the neuraminidase (NA) of H5N1 influenza A virus markedly deglycosylates and degrades lysosome-associated membrane proteins (LAMPs; the most abundant membrane proteins of lysosome), which induces lysosomal rupture, and finally leads to cell death of alveolar epithelial carcinoma A549 cells and human tracheal epithelial cells. The NA inhibitors peramivir and zanamivir could effectively block the deglycosylation of LAMPs, inhibit the virus cell entry, and prevent cell death induced by the H5N1 influenza virus. The NA of seasonal H1N1 virus, however, does not share these characteristics. Our findings not only reveal a novel role of NA in the early stage of the H5N1 influenza virus life cycle but also elucidate the molecular mechanism of lysosomal rupture crucial for influenza virus induced cell death.

Importance: The integrity of lysosomes is vital for maintaining cell homeostasis, cellular defense and clearance of invading pathogens. This study shows that the H5N1 influenza virus could induce lysosomal rupture through deglycosylating lysosome-associated membrane proteins (LAMPs) mediated by the neuraminidase activity of NA protein. NA inhibitors such as peramivir and zanamivir could inhibit the deglycosylation of LAMPs and protect lysosomes, which also further interferes with the H5N1 influenza virus infection at early stage of life cycle. This work is significant because it presents new concepts for NA's function, as well as for influenza inhibitors' mechanism of action, and could partially explain the high mortality and high viral load after H5N1 virus infection in human beings and why NA inhibitors have more potent therapeutic effects for lethal avian influenza virus infections at early stage.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Acids, Carbocyclic
  • Cell Death / drug effects
  • Cell Line, Tumor
  • Cell Membrane / chemistry
  • Cell Membrane / enzymology*
  • Cyclopentanes / pharmacology
  • Cytosol / drug effects
  • Cytosol / enzymology
  • Cytosol / virology
  • Enzyme Inhibitors / pharmacology
  • Epithelial Cells / drug effects
  • Epithelial Cells / virology
  • Guanidines / pharmacology
  • Humans
  • Hydrolysis
  • Influenza A Virus, H1N1 Subtype / chemistry
  • Influenza A Virus, H1N1 Subtype / enzymology
  • Influenza A Virus, H5N1 Subtype / chemistry
  • Influenza A Virus, H5N1 Subtype / enzymology
  • Lysosomal Membrane Proteins / chemistry
  • Lysosomal Membrane Proteins / metabolism*
  • Lysosomes / drug effects
  • Lysosomes / enzymology*
  • Lysosomes / virology
  • Neuraminidase / metabolism*
  • Protein Binding
  • Proteolysis
  • Respiratory Mucosa / drug effects
  • Respiratory Mucosa / virology
  • Species Specificity
  • Viral Proteins / metabolism*
  • Virus Internalization / drug effects
  • Zanamivir / pharmacology

Substances

  • Acids, Carbocyclic
  • Cyclopentanes
  • Enzyme Inhibitors
  • Guanidines
  • Lysosomal Membrane Proteins
  • Viral Proteins
  • Neuraminidase
  • Zanamivir
  • peramivir