Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Pathog Immun ; 4(2): 196-234, 2019.
Article in English | MEDLINE | ID: mdl-31583330

ABSTRACT

BACKGROUND: Human papillomavirus (HPV) is linked to nearly all cases of cervical cancer. Despite available vaccines, a deeper understanding of the immune response to HPV is needed. Human α-defensin 5 (HD5), an innate immune effector peptide, blocks infection of multiple sero-types of HPV, including high-risk HPV16. While a common mechanism of α-defensin anti-viral activity against nonenveloped viruses such as HPV has emerged, there is limited understanding of how α-defensins bind to viral capsids to block infection. METHODS: We have used cryo-electron microscopy (cryoEM), mass spectrometry (MS) crosslinking and differential lysine modification studies, and molecular dynamics (MD) simulations to probe the interaction of HPV16 pseudovirions (PsVs) with HD5. RESULTS: CryoEM single particle reconstruction did not reveal HD5 density on the capsid surface. Rather, increased density was observed under the capsid shell in the presence of HD5. MS studies indicate that HD5 binds near the L1 and L2 capsid proteins and specifically near the C-terminal region of L1. MD simulations indicate that favorable electrostatic interactions can be formed between HD5 and the L1 C-terminal tail. CONCLUSIONS: A model is presented for how HD5 affects HPV16 structure and cell entry. In this model, HD5 binds to disordered regions of L1 and L2 protruding from the icosahedrally ordered capsid. HD5 acts to cement interactions between L1 and L2 and leads to a closer association of the L2/genome core with the L1 capsid. This model provides a structural rationale for our prior observation that HD5 interferes with the separation of L1 from the L2/genome complex during cell entry.

2.
PLoS Pathog ; 13(6): e1006446, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28622386

ABSTRACT

The small intestinal epithelium produces numerous antimicrobial peptides and proteins, including abundant enteric α-defensins. Although they most commonly function as potent antivirals in cell culture, enteric α-defensins have also been shown to enhance some viral infections in vitro. Efforts to determine the physiologic relevance of enhanced infection have been limited by the absence of a suitable cell culture system. To address this issue, here we use primary stem cell-derived small intestinal enteroids to examine the impact of naturally secreted α-defensins on infection by the enteric mouse pathogen, mouse adenovirus 2 (MAdV-2). MAdV-2 infection was increased when enteroids were inoculated across an α-defensin gradient in a manner that mimics oral infection but not when α-defensin levels were absent or bypassed through other routes of inoculation. This increased infection was a result of receptor-independent binding of virus to the cell surface. The enteroid experiments accurately predicted increased MAdV-2 shedding in the feces of wild type mice compared to mice lacking functional α-defensins. Thus, our studies have shown that viral infection enhanced by enteric α-defensins may reflect the evolution of some viruses to utilize these host proteins to promote their own infection.


Subject(s)
Adenoviridae Infections/virology , Adenoviridae/physiology , Intestine, Small/metabolism , alpha-Defensins/metabolism , Adenoviridae/genetics , Animals , Female , Host-Pathogen Interactions , Humans , Intestine, Small/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Virus Shedding , alpha-Defensins/genetics
3.
mBio ; 8(1)2017 01 24.
Article in English | MEDLINE | ID: mdl-28119475

ABSTRACT

α-Defensins are an important class of abundant innate immune effectors that are potently antiviral against a number of nonenveloped viral pathogens; however, a common mechanism to explain their ability to block infection by these unrelated viruses is lacking. We previously found that human defensin 5 (HD5) blocks a critical host-mediated proteolytic processing step required for human papillomavirus (HPV) infection. Here, we show that bypassing the requirement for this cleavage failed to abrogate HD5 inhibition. Instead, HD5 altered HPV trafficking in the cell. In the presence of an inhibitory concentration of HD5, HPV was internalized and reached the early endosome. The internalized capsid became permeable to antibodies and proteases; however, HD5 prevented dissociation of the viral capsid from the genome, reduced viral trafficking to the trans-Golgi network, redirected the incoming viral particle to the lysosome, and accelerated the degradation of internalized capsid proteins. This mechanism is equivalent to the mechanism by which HD5 inhibits human adenovirus. Thus, our data support capsid stabilization and redirection to the lysosome during infection as a general antiviral mechanism of α-defensins against nonenveloped viruses. IMPORTANCE: Although the antiviral activity of α-defensins against enveloped viruses can be largely explained by interference with receptor binding and fusion, a common mechanism for inhibition of nonenveloped viruses remains elusive. In studies of a prominent human α-defensin that is expressed in the gut and in the male and female genitourinary tract, we discovered striking parallels between the mechanisms of inhibition of HPV and human adenovirus infection. Thus, detailed studies of the impact of α-defensins on the intracellular trafficking of two disparate viruses support a general mechanism of α-defensin antiviral activity against nonenveloped viruses.


Subject(s)
Capsid Proteins/metabolism , Human papillomavirus 16/immunology , Lysosomes/metabolism , alpha-Defensins/metabolism , Cell Line , Humans , Proteolysis
4.
J Virol ; 90(11): 5216-5218, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27009960

ABSTRACT

Defensins are innate immune effector peptides expressed at mucosal surfaces throughout the human body and are potently antiviral in vitro The role of defensins in viral pathogenesis in vivo is poorly understood; however, recent studies have revealed that defensin-virus interactions in vivo are complicated and distinct from their proposed antiviral mechanisms in vitro These findings highlight the need for additional research that connects defensin neutralization of viruses in cell culture to in vivo antiviral mechanisms.


Subject(s)
Defensins/metabolism , Immunomodulation , Mucous Membrane/immunology , Mucous Membrane/virology , Virus Diseases/immunology , Viruses/immunology , Animals , Antiviral Agents/immunology , Antiviral Agents/metabolism , Defensins/immunology , Humans , Mucous Membrane/chemistry , Mucous Membrane/physiology , Virus Diseases/virology , Viruses/metabolism , alpha-Defensins/immunology , alpha-Defensins/metabolism
5.
PLoS Pathog ; 12(3): e1005474, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26933888

ABSTRACT

α-defensins are abundant antimicrobial peptides with broad, potent antibacterial, antifungal, and antiviral activities in vitro. Although their contribution to host defense against bacteria in vivo has been demonstrated, comparable studies of their antiviral activity in vivo are lacking. Using a mouse model deficient in activated α-defensins in the small intestine, we show that Paneth cell α-defensins protect mice from oral infection by a pathogenic virus, mouse adenovirus 1 (MAdV-1). Survival differences between mouse genotypes are lost upon parenteral MAdV-1 infection, strongly implicating a role for intestinal defenses in attenuating pathogenesis. Although differences in α-defensin expression impact the composition of the ileal commensal bacterial population, depletion studies using broad-spectrum antibiotics revealed no effect of the microbiota on α-defensin-dependent viral pathogenesis. Moreover, despite the sensitivity of MAdV-1 infection to α-defensin neutralization in cell culture, we observed no barrier effect due to Paneth cell α-defensin activation on the kinetics and magnitude of MAdV-1 dissemination to the brain. Rather, a protective neutralizing antibody response was delayed in the absence of α-defensins. This effect was specific to oral viral infection, because antibody responses to parenteral or mucosal ovalbumin exposure were not affected by α-defensin deficiency. Thus, α-defensins play an important role as adjuvants in antiviral immunity in vivo that is distinct from their direct antiviral activity observed in cell culture.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/immunology , Anti-Infective Agents/immunology , Antibodies, Neutralizing/immunology , Antiviral Agents/immunology , Defensins/immunology , Animals , Female , Humans , Ileum/immunology , Intestine, Small/immunology , Intestines/immunology , Male , Mice , Mice, Inbred C57BL , Paneth Cells/immunology , alpha-Defensins/immunology
6.
J Virol ; 89(5): 2866-74, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25540379

ABSTRACT

UNLABELLED: Human papillomavirus (HPV) is a significant oncogenic virus, but the innate immune response to HPV is poorly understood. Human α-defensin 5 (HD5) is an innate immune effector peptide secreted by epithelial cells in the genitourinary tract. HD5 is broadly antimicrobial, exhibiting potent antiviral activity against HPV at physiologic concentrations; however, the specific mechanism of HD5-mediated inhibition against HPV is unknown. During infection, the HPV capsid undergoes several critical cell-mediated viral protein processing steps, including unfolding and cleavage of the minor capsid protein L2 by host cyclophilin B and furin. Using HPV16 pseudovirus, we show that HD5 interacts directly with the virus and inhibits the furin-mediated cleavage of L2 at the cell surface during infection at a step downstream of the cyclophilin B-mediated unfolding of L2. Importantly, HD5 does not affect the enzymatic activity of furin directly. Thus, our data support a model in which HD5 prevents furin from accessing L2 by occluding the furin cleavage site via direct binding to the viral capsid. IMPORTANCE: Our study elucidates a new antiviral action for α-defensins against nonenveloped viruses in which HD5 directly interferes with a critical host-mediated viral processing step, furin cleavage of L2, at the cell surface. Blocking this key event has deleterious effects on the intracellular steps of virus infection. Thus, in addition to informing the antiviral mechanisms of α-defensins, our studies highlight the critical role of furin cleavage in HPV entry. Innate immune control, mediated in part by α-defensins expressed in the genital mucosa, may influence susceptibility to HPV infections that lead to cervical cancer. Moreover, understanding the mechanism of these natural antivirals may inform the design of therapeutics to limit HPV infection.


Subject(s)
Capsid Proteins/metabolism , Furin/antagonists & inhibitors , Human papillomavirus 16/immunology , Human papillomavirus 16/physiology , Oncogene Proteins, Viral/metabolism , Virus Internalization , alpha-Defensins/metabolism , Cell Line , Humans , Immunity, Innate
7.
PLoS Pathog ; 10(9): e1004360, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25188351

ABSTRACT

Human α-defensins are potent anti-microbial peptides with the ability to neutralize bacterial and viral targets. Single alanine mutagenesis has been used to identify determinants of anti-bacterial activity and binding to bacterial proteins such as anthrax lethal factor. Similar analyses of α-defensin interactions with non-enveloped viruses are limited. We used a comprehensive set of human α-defensin 5 (HD5) and human neutrophil peptide 1 (HNP1) alanine scan mutants in a combination of binding and neutralization assays with human adenovirus (AdV) and human papillomavirus (HPV). We have identified a core of critical hydrophobic residues that are common determinants for all of the virus-defensin interactions that were analyzed, while specificity in viral recognition is conferred by specific surface-exposed charged residues. The hydrophobic residues serve multiple roles in maintaining the tertiary and quaternary structure of the defensins as well as forming an interface for virus binding. Many of the important solvent-exposed residues of HD5 group together to form a critical surface. However, a single discrete binding face was not identified for HNP1. In lieu of whole AdV, we used a recombinant capsid subunit comprised of penton base and fiber in quantitative binding studies and determined that the anti-viral potency of HD5 was a function of stoichiometry rather than affinity. Our studies support a mechanism in which α-defensins depend on hydrophobic and charge-charge interactions to bind at high copy number to these non-enveloped viruses to neutralize infection and provide insight into properties that guide α-defensin anti-viral activity.


Subject(s)
Adenovirus Infections, Human/prevention & control , Adenoviruses, Human/drug effects , Papillomaviridae/drug effects , Papillomavirus Infections/prevention & control , alpha-Defensins/chemistry , alpha-Defensins/pharmacology , Adenovirus Infections, Human/virology , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Mutagenesis , Papillomavirus Infections/virology , Protein Conformation , Surface Plasmon Resonance , Virus Attachment
9.
J Mol Biol ; 425(24): 4965-80, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24095897

ABSTRACT

Defensins are an effector component of the innate immune system with broad antimicrobial activity. Humans express two types of defensins, α- and ß-defensins, which have antiviral activity against both enveloped and non-enveloped viruses. The diversity of defensin-sensitive viral species reflects a multitude of antiviral mechanisms. These include direct defensin targeting of viral envelopes, glycoproteins, and capsids in addition to inhibition of viral fusion and post-entry neutralization. Binding and modulation of host cell surface receptors and disruption of intracellular signaling by defensins can also inhibit viral replication. In addition, defensins can function as chemokines to augment and alter adaptive immune responses, revealing an indirect antiviral mechanism. Nonetheless, many questions regarding the antiviral activities of defensins remain. Although significant mechanistic data are known for α-defensins, molecular details for ß-defensin inhibition are mostly lacking. Importantly, the role of defensin antiviral activity in vivo has not been addressed due to the lack of a complete defensin knockout model. Overall, the antiviral activity of defensins is well established as are the variety of mechanisms by which defensins achieve this inhibition; however, additional research is needed to fully understand the role of defensins in viral pathogenesis.


Subject(s)
Antiviral Agents/metabolism , Defensins/physiology , Immunity, Innate/immunology , Signal Transduction/immunology , Viruses/immunology , Antiviral Agents/immunology , Defensins/genetics , Humans , Models, Immunological , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Virus Replication , alpha-Defensins/genetics , alpha-Defensins/metabolism , alpha-Defensins/physiology , beta-Defensins/genetics , beta-Defensins/metabolism , beta-Defensins/physiology
10.
J Biol Chem ; 287(29): 24554-62, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22637473

ABSTRACT

Human α-defensins, such as human α-defensin 5 (HD5), block infection of non-enveloped viruses, including human adenoviruses (AdV), papillomaviruses (HPV), and polyomaviruses. Through mutational analysis of HD5, we have identified arginine residues that contribute to antiviral activity against AdV and HPV. Of two arginine residues paired on one face of HD5, Arg-28 is critical for both viruses, while Arg-9 is only important for AdV. Two arginine residues on the opposite face of the molecule (Arg-13 and Arg-32) and unpaired Arg-25 are less important for both. In addition, hydrophobicity at residue 29 is a major determinant of anti-adenoviral activity, and a chemical modification that prevents HD5 self-association was strongly attenuating. Although HD5 binds to the capsid of AdV, the molecular basis for this interaction is undefined. Capsid binding by HD5 is not purely charge-dependent, as substitution of lysine for Arg-9 and Arg-28 was deleterious. Analysis of HD5 analogs that retained varying levels of potency demonstrated that anti-adenoviral activity is directly correlated with HD5 binding to the virus, confirming that the viral capsid rather than the cell is the relevant target. Also, AdV aggregation induced by HD5 binding is not sufficient for neutralization. Rather, these studies confirm that the major mechanism of HD5-mediated neutralization of AdV depends upon specific binding to the viral capsid through interactions mediated in part by critical arginine residues, hydrophobicity at residue 29, and multimerization of HD5, which increases initial binding of virus to the cell but prevents subsequent viral uncoating and genome delivery to the nucleus.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Arginine/chemistry , Viruses/drug effects , alpha-Defensins/chemistry , alpha-Defensins/pharmacology , Cell Line, Tumor , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Protein Multimerization , Structure-Activity Relationship
11.
PLoS One ; 7(3): e32661, 2012.
Article in English | MEDLINE | ID: mdl-22438882

ABSTRACT

Retinoic acid inducible gene-I (RIG-I) is a key regulator of antiviral immunity. RIG-I is generally thought to be activated by ssRNA species containing a 5'-triphosphate (PPP) group or by unphosphorylated dsRNA up to ~300 bp in length. However, it is not yet clear how changes in the length, nucleotide sequence, secondary structure, and 5' end modification affect the abilities of these ligands to bind and activate RIG-I. To further investigate these parameters in the context of naturally occurring ligands, we examined RNA sequences derived from the 5' and 3' untranslated regions (UTR) of the influenza virus NS1 gene segment. As expected, RIG-I-dependent interferon-ß (IFN-ß) induction by sequences from the 5' UTR of the influenza cRNA or its complement (26 nt in length) required the presence of a 5'PPP group. In contrast, activation of RIG-I by the 3' UTR cRNA sequence or its complement (172 nt) exhibited only a partial 5'PPP-dependence, as capping the 5' end or treatment with CIP showed a modest reduction in RIG-I activation. Furthermore, induction of IFN-ß by a smaller, U/A-rich region within the 3' UTR was completely 5'PPP-independent. Our findings demonstrated that RNA sequence, length, and secondary structure all contributed to whether or not the 5'PPP moiety is needed for interferon induction by RIG-I.


Subject(s)
DEAD-box RNA Helicases/metabolism , Genome, Viral , Influenza A virus/genetics , Influenza A virus/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , 3' Untranslated Regions , 5' Untranslated Regions , Binding Sites/genetics , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases/chemistry , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , Influenza A virus/immunology , Interferon-beta/biosynthesis , Interferon-beta/genetics , Kinetics , Ligands , Nucleic Acid Conformation , RNA, Viral/chemistry , Receptors, Immunologic , Viral Nonstructural Proteins/genetics
12.
Virol J ; 7: 102, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20492658

ABSTRACT

BACKGROUND: Emergence of drug-resistant strains of influenza viruses, including avian H5N1 with pandemic potential, 1918 and 2009 A/H1N1 pandemic viruses to currently used antiviral agents, neuraminidase inhibitors and M2 Ion channel blockers, underscores the importance of developing novel antiviral strategies. Activation of innate immune pathogen sensor Retinoic Acid Inducible Gene-I (RIG-I) has recently been shown to induce antiviral state. RESULTS: In the present investigation, using real time RT-PCR, immunofluorescence, immunoblot, and plaque assay we show that 5'PPP-containing single stranded RNA (5'PPP-RNA), a ligand for the intracytoplasmic RNA sensor, RIG-I can be used as a prophylactic agent against known drug-resistant avian H5N1 and pandemic influenza viruses. 5'PPP-RNA treatment of human lung epithelial cells inhibited replication of drug-resistant avian H5N1 as well as 1918 and 2009 pandemic influenza viruses in a RIG-I and type 1 interferon dependant manner. Additionally, 5'PPP-RNA treatment also inhibited 2009 H1N1 viral replication in vivo in mice. CONCLUSIONS: Our findings suggest that 5'PPP-RNA mediated activation of RIG-I can suppress replication of influenza viruses irrespective of their genetic make-up, pathogenicity, and drug-sensitivity status.


Subject(s)
DEAD-box RNA Helicases/metabolism , Disease Outbreaks , Drug Resistance, Viral , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/drug effects , Influenza, Human/virology , RNA, Viral/metabolism , Virus Replication , Animals , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/physiology , Influenza, Human/epidemiology , Influenza, Human/genetics , Mice , Mice, Inbred BALB C , RNA, Viral/chemical synthesis , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/pharmacology , Receptors, Immunologic
SELECTION OF CITATIONS
SEARCH DETAIL
...