Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
2.
Biol Cell ; 114(7): 185-198, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35389514

ABSTRACT

MAGI-1 is a critical cellular scaffolding protein with over 110 different cellular and microbial protein interactors. Since the discovery of MAGI-1 in 1997, MAGI-1 has been implicated in diverse cellular functions such as polarity, cell-cell communication, neurological processes, kidney function, and a host of diseases including cancer and microbial infection. Additionally, MAGI-1 has undergone nomenclature changes in response to the discovery of an additional PDZ domain, leading to lack of continuity in the literature. We address the nomenclature of MAGI-1 as well as summarize many of the critical functions of the known interactions. Given the importance of many of the interactors, such as human papillomavirus E6, the Coxsackievirus and adenovirus receptor (CAR), and PTEN, the enhancement or disruption of MAGI-based interactions has the potential to affect cellular functions that can potentially be harnessed as a therapeutic strategy for a variety of diseases.


Subject(s)
PDZ Domains , Humans
3.
Pathogens ; 11(2)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35215116

ABSTRACT

The coxsackievirus and adenovirus receptor (CAR) is an essential cellular protein that is involved in cell adhesion, cell signaling, and viral infection. The 8-exon encoded isoform (CAREx8) resides at the apical surface of polarized epithelia, where it is accessible as a receptor for adenovirus entering the airway lumen. Given its pivotal role in viral infection, it is a target for antiviral strategies. To understand the regulation of CAREx8 and determine the feasibility of receptor downregulation, the half-life of total and apical localized CAREx8 was determined and correlated with adenovirus transduction. Total and apical CAREx8 has a relatively short half-life of approximately 2 h. The half-life of apical CAREx8 correlates well with adenovirus transduction. These results suggest that antiviral strategies that aim to degrade the primary receptor for apical adenovirus infection will be effective within a relatively short time frame after application.

4.
J Virol ; 95(13): e0004621, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33762416

ABSTRACT

Adenoviruses (AdVs) are etiological agents of gastrointestinal, heart, eye, and respiratory tract infections that can be lethal for immunosuppressed people. Many AdVs use the coxsackievirus and adenovirus receptor (CAR) as a primary receptor. The CAR isoform resulting from alternative splicing that includes the eighth exon, CAREx8, localizes to the apical surface of polarized epithelial cells and is responsible for the initiation of AdV infection. We have shown that the membrane level of CAREx8 is tightly regulated by two MAGI-1 PDZ domains, PDZ2 and PDZ4, resulting in increased or decreased AdV transduction, respectively. We hypothesized that targeting the interactions between the MAGI-1 PDZ2 domain and CAREx8 would decrease the apical CAREx8 expression level and prevent AdV infection. Decoy peptides that target MAGI-1 PDZ2 were synthesized (TAT-E6 and TAT-NET1). PDZ2 binding peptides decreased CAREx8 expression and reduced AdV transduction. CAREx8 degradation was triggered by the activation of the regulated intramembrane proteolysis (RIP) pathway through a disintegrin and metalloproteinase (ADAM17) and γ-secretase. Further analysis revealed that ADAM17 interacts directly with the MAGI-1 PDZ3 domain, and blocking the PDZ2 domain enhanced the accessibility of ADAM17 to the substrate (CAREx8). Finally, we validated the efficacy of TAT-PDZ2 peptides in protecting the epithelia from AdV transduction in vivo using a novel transgenic animal model. Our data suggest that TAT-PDZ2 binding peptides are novel anti-AdV molecules that act by enhanced RIP of CAREx8 and decreased AdV entry. This strategy has additional translational potential for targeting other viral receptors that have PDZ binding domains, such as the angiotensin-converting enzyme 2 receptor. IMPORTANCE Adenovirus is a common threat in immunosuppressed populations and military recruits. There are no currently approved treatments/prophylactic agents that protect from most AdV infections. Here, we developed peptide-based small molecules that can suppress AdV infection of polarized epithelia by targeting the AdV receptor, coxsackievirus and adenovirus receptor (CAREx8). The newly discovered peptides target a specific PDZ domain of the CAREx8-interacting protein MAGI-1 and decrease AdV transduction in multiple polarized epithelial models. Peptide-induced CAREx8 degradation is triggered by extracellular domain (ECD) shedding through ADAM17 followed by γ-secretase-mediated nuclear translocation of the C-terminal domain. The enhanced shedding of the CAREx8 ECD further protected the epithelium from AdV infection. Taken together, these novel molecules protect the epithelium from AdV infection. This approach may be applicable to the development of novel antiviral molecules against other viruses that use a receptor with a PDZ binding domain.


Subject(s)
ADAM17 Protein/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adenoviridae Infections/prevention & control , Cell Adhesion Molecules/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/antagonists & inhibitors , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Guanylate Kinases/metabolism , 3T3 Cells , Adenoviridae/immunology , Amyloid Precursor Protein Secretases/metabolism , Animals , Dogs , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Mice , Protein Domains
5.
Viruses ; 14(1)2021 12 22.
Article in English | MEDLINE | ID: mdl-35062217

ABSTRACT

Human adenoviruses (HAdV) cause a variety of infections in human hosts, from self-limited upper respiratory tract infections in otherwise healthy people to fulminant pneumonia and death in immunocompromised patients. Many HAdV enter polarized epithelial cells by using the primary receptor, the Coxsackievirus and adenovirus receptor (CAR). Recently published data demonstrate that a potent neutrophil (PMN) chemoattractant, interleukin-8 (IL-8), stimulates airway epithelial cells to increase expression of the apical isoform of CAR (CAREx8), which results in increased epithelial HAdV type 5 (HAdV5) infection. However, the mechanism for PMN-enhanced epithelial HAdV5 transduction remains unclear. In this manuscript, the molecular mechanisms behind PMN mediated enhancement of epithelial HAdV5 transduction are characterized using an MDCK cell line that stably expresses human CAREx8 under a doxycycline inducible promoter (MDCK-CAREx8 cells). Contrary to our hypothesis, PMN exposure does not enhance HAdV5 entry by increasing CAREx8 expression nor through activation of non-specific epithelial endocytic pathways. Instead, PMN serine proteases are responsible for PMN-mediated enhancement of HAdV5 transduction in MDCK-CAREx8 cells. This is evidenced by reduced transduction upon inhibition of PMN serine proteases and increased transduction upon exposure to exogenous human neutrophil elastase (HNE). Furthermore, HNE exposure activates epithelial autophagic flux, which, even when triggered through other mechanisms, results in a similar enhancement of epithelial HAdV5 transduction. Inhibition of F-actin with cytochalasin D partially attenuates PMN mediated enhancement of HAdV transduction. Taken together, these findings suggest that HAdV5 can leverage innate immune responses to establish infections.


Subject(s)
Adenoviruses, Human/pathogenicity , Epithelial Cells/virology , Leukocyte Elastase/metabolism , Neutrophils/immunology , Virus Internalization , Adenoviruses, Human/immunology , Adenoviruses, Human/physiology , Animals , Autophagy , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Cytochalasin B/pharmacology , Dogs , Endocytosis , Humans , Immunity, Innate , Macrolides/pharmacology , Madin Darby Canine Kidney Cells , Receptors, Virus/metabolism
6.
MethodsX ; 7: 101149, 2020.
Article in English | MEDLINE | ID: mdl-33304833

ABSTRACT

CRISPR-Cas9 gene editing has made it possible to specifically edit genes in a myriad of target cells. Here, a method for isoform-specific editing and clonal selection in Madin-Darby canine kidney (MDCK) epithelial cells is described in detail. This approach was used to address a long-standing question in virology of how adenovirus enters polarized epithelia from the apical surface. Our method relies on selecting two sgRNA sequences, cloning them into a suitable fluorescently labeled Cas9 vector system, and subsequently transfecting our MDCK epithelium and selecting isoform-specific Coxsackievirus and adenovirus receptor knockout clones. Utilization of this method is readily applicable to many other genetic targets in epithelial cells.•Simultaneous utilization of an sgRNA upstream and an sgRNA downstream of a target sequence allows for deletion of the intervening sequence, including whole exons.•Sorting of cells positive for fluorescent marker gene expression enhances the identification of partial and biallelic gene knockout.•PCR screening allows relatively fast and efficient determination of isoform-specific deletion.

7.
FEBS Lett ; 594(12): 1828-1837, 2020 06.
Article in English | MEDLINE | ID: mdl-32298477

ABSTRACT

The coxsackievirus and adenovirus receptor (CAR) is an essential multifunctional cellular protein that is only beginning to be understood. CAR serves as a receptor for many adenoviruses, human group B coxsackieviruses, swine vesicular disease virus, and possibly other viruses. While named for its function as a viral receptor, CAR is also involved in cell adhesion, immune cell activation, synaptic transmission, and signaling. Knockout mouse models were first to identify some of these biological functions; however, tissue-specific model systems have shed light on the complexity of different CAR isoforms and their specific activities. Many of these functions are mediated by the large number of interacting proteins described so far, and several new putative interactions have recently been discovered. As antiviral and gene therapy strategies that target CAR continue to emerge, future work poised to understand the biological implications of manipulating CAR in vivo is critical.


Subject(s)
Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Adenoviridae Infections/metabolism , Alternative Splicing , Animals , Gene Expression Regulation, Developmental , Humans , Mice , Protein Isoforms
8.
Virology ; 536: 20-26, 2019 10.
Article in English | MEDLINE | ID: mdl-31394408

ABSTRACT

The Coxsackievirus and adenovirus receptor (CAR) is both a viral receptor and cell adhesion protein. CAR has two transmembrane isoforms that localize distinctly in polarized epithelial cells. Whereas the seven exon-encoded isoform (CAREx7) exhibits basolateral localization, the eight exon-encoded isoform (CAREx8) can localize to the apical epithelial surface where it can mediate luminal adenovirus infection. To further understand the distinct biological functions of these two isoforms, CRISPR/Cas9 genomic editing was used to specifically delete the eighth exon of the CXADR gene in a Madine Darby Canine Kidney (MDCK) cell line with a stably integrated lentiviral doxycycline-inducible CAREx8 cDNA. The gene-edited clone demonstrated a significant reduction in adenovirus susceptibility when both partially and fully polarized, and doxycycline-induction of CAREx8 restored sensitivity to adenovirus. These data reinforce the importance of CAREx8 in apical adenovirus infection and provide a new model cell line to probe isoform specific biological functions of CAR.


Subject(s)
Adenoviruses, Human/genetics , CRISPR-Cas Systems , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Gene Editing/methods , Gene Expression Regulation, Viral , Adenoviruses, Human/metabolism , Animals , Base Sequence , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Dogs , Doxycycline/pharmacology , Exons , Humans , Madin Darby Canine Kidney Cells , Promoter Regions, Genetic/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
9.
J Invest Dermatol ; 139(2): 448-454, 2019 02.
Article in English | MEDLINE | ID: mdl-30227140

ABSTRACT

UVB wavelengths of light induce the formation of photoproducts in genomic DNA that are potentially mutagenic and detrimental to epidermal cell function. The mineralocorticoid and androgen receptor antagonist spironolactone (SP) was recently identified as an inhibitor of UV photoproduct removal in human cancer cells in vitro via its ability to promote the rapid proteolytic degradation of the DNA repair protein XPB. Using normal human keratinocytes in vitro and skin explants ex vivo, we found that SP rapidly depleted XPB protein in both systems and abrogated two major responses to UVB-induced DNA damage, including the removal of UV photoproducts from genomic DNA and the activation of ATR/ATM DNA damage kinase signaling. These effects were also correlated with both mutagenesis and a predisposition to UVB-induced cell death but were unique to SP, because neither the SP metabolites canrenone and 7α-thiomethylspironolactone nor the more specific mineralocorticoid receptor antagonist eplerenone affected XPB protein levels or the UVB response. Our findings provide an approach for studying XPB and its roles in the UVB DNA damage response in human skin ex vivo and indicate that SP may increase UVB mutagenesis and skin cancer risk in certain individuals.


Subject(s)
DNA Helicases/antagonists & inhibitors , DNA Repair/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Mutagenesis/drug effects , Spironolactone/toxicity , Cells, Cultured , DNA Damage/radiation effects , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Humans , Keratinocytes , Mutagenesis/radiation effects , Primary Cell Culture , Skin/drug effects , Skin/pathology , Skin/radiation effects , Ultraviolet Rays/adverse effects
10.
J Virol Methods ; 250: 47-54, 2017 12.
Article in English | MEDLINE | ID: mdl-28918073

ABSTRACT

A directed evolution approach was used to select for Adeno-associated virus (AAV) capsids that would exhibit more tropism toward an HIV-1 producer T cell line with the long-term goal of developing improved gene transfer vectors. A library of AAV variants was used to infect H9 T cells previously infected or uninfected by HIV-1 followed by AAV amplification with wild-type adenovirus. Six rounds of biological selection were performed, including negative selection and diversification after round three. The H9 T cells were successfully infected with all three wild-type viruses (AAV, adenovirus, and HIV-1). Four AAV cap mutants best representing the small number of variants emerging after six rounds of selection were chosen for further study. These mutant capsids were used to package an AAV vector and subsequently used to infect H9 cells that were previously infected or uninfected by HIV-1. A quantitative polymerase chain reaction assay was performed to measure cell-associated AAV genomes. Two of the four cap mutants showed a significant increase in the amount of cell-associated genomes as compared to wild-type AAV2. This study shows that directed evolution can be performed successfully to select for mutants with improved tropism for a T cell line in the presence of HIV-1.


Subject(s)
Capsid , Dependovirus/genetics , Dependovirus/isolation & purification , Directed Molecular Evolution/methods , T-Lymphocytes/virology , Cell Line , Gene Library , Gene Transfer Techniques , Genetic Vectors , HIV-1/genetics , HIV-1/physiology , HeLa Cells , Humans , Mutation , Viral Tropism , Virus Uncoating
11.
Virus Res ; 242: 1-6, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28870470

ABSTRACT

The human poliovirus receptor (PVR) is a cell surface protein with a multitude of functions in human biology. PVR was initially identified as the receptor for the human poliovirus and recent discoveries have given a greater insight into both its morphology and its function. Alternative splicing of the PVR gene results in a total of 4 alternatively spliced isoforms. Two of these isoforms lack a complete transmembrane domain and are considered soluble and block viral infection; the remaining two transmembrane isoforms differ only at their extreme C-terminal domains resulting in differential localization in epithelia and polarity of viral infection. In addition to its role as a receptor for the human poliovirus, several native biological functions have also been uncovered. PVR is an important cell adhesion protein and is involved in the transendothelial migration of leukocytes. Through its interactions with CD226 and TIGIT, transmembrane proteins found on leukocytes, PVR is a key regulator of the cell-mediated immune response. As PVR is differentially regulated in a broad spectrum of cancers, it has a strong potential for clinical use as a biomarker. PVR is also a possible target for novel cancer therapies. Utilizing its natural tropism for PVR, a genetically modified form of the live attenuated poliovirus vaccine is currently being tested for its ability to locate and destroy certain tumors. These recent studies emphasize the importance of PVR in human biology and demonstrate its utility beyond being a viral receptor protein.


Subject(s)
Protein Isoforms/metabolism , Receptors, Virus/metabolism , Cell Adhesion , Cell Movement , Humans , Immunity, Cellular , Leukocytes/immunology , Protein Isoforms/genetics , RNA Splicing , Receptors, Virus/genetics
12.
Virology ; 502: 144-151, 2017 02.
Article in English | MEDLINE | ID: mdl-28049062

ABSTRACT

The abundance and accessibility of a primary virus receptor are critical factors that impact the susceptibility of a host cell to virus infection. The Coxsackievirus and adenovirus receptor (CAR) has two transmembrane isoforms that occur due to alternative splicing and differ in localization and function in polarized epithelia. To determine the relevance of isoform-specific expression across cell types, the abundance and localization of both isoforms were determined in ten common cell lines, and correlated with susceptibility to adenovirus transduction relative to polarized primary human airway epithelia. Data show that the gene and protein expression for each isoform of CAR varies significantly between cell lines and polarization, as indicated by high transepithelial resistance, is inversely related to adenovirus transduction. In summary, the variability of polarity and isoform-specific expression among model cells are critical parameters that must be considered when evaluating the clinical relevance of potential adenovirus-mediated gene therapy and anti-adenovirus strategies.


Subject(s)
Adenoviridae Infections/metabolism , Adenoviridae/genetics , Epithelial Cells/virology , Receptors, Virus/metabolism , Transduction, Genetic , Adenoviridae/physiology , Adenoviridae Infections/genetics , Adenoviridae Infections/physiopathology , Adenoviridae Infections/virology , Cell Line , Cell Polarity , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Virus/genetics , Respiratory System/cytology , Respiratory System/metabolism , Respiratory System/virology
13.
Hum Gene Ther Methods ; 27(6): 219-227, 2016 12.
Article in English | MEDLINE | ID: mdl-27820963

ABSTRACT

Adipose-derived stem cells (ASCs) have shown potential in the treatment of a myriad of diseases; however, infusion of cells alone is unlikely to provide the full range of potential therapeutic applications. Transient genetic manipulation of ASCs could increase their repair and regeneration characteristics in a disease-specific context, essentially transforming them into drug-eluting depots. The goal of this study was to determine the optimal parameters necessary to transduce ASCs with recombinant adeno-associated virus (rAAV), an approved gene therapy vector that has never been associated with disease. Transduction and duration of gene expression of the most common recombinant AAV vectors were tested in this study. Among all tested serotypes, rAAV5 resulted in both the highest and longest term expression. Furthermore, we determined the glycosylation profile of ASCs before and after neuraminidase treatment and demonstrate that rAAV5 transduction requires plasma membrane-associated sialic acid. Future studies will focus on the optimization of gene delivery to ASCs, using rAAV5 as the vector of choice, to drive biological drug delivery, engraftment, and disease correction.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Stem Cells , Transduction, Genetic , Adipose Tissue/cytology , Animals , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Serogroup
14.
PLoS Pathog ; 11(3): e1004696, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25768646

ABSTRACT

Prevention of viral-induced respiratory disease begins with an understanding of the factors that increase or decrease susceptibility to viral infection. The primary receptor for most adenoviruses is the coxsackievirus and adenovirus receptor (CAR), a cell-cell adhesion protein normally localized at the basolateral surface of polarized epithelia and involved in neutrophil transepithelial migration. Recently, an alternate isoform of CAR, CAREx8, has been identified at the apical surface of polarized airway epithelia and is implicated in viral infection from the apical surface. We hypothesized that the endogenous role of CAREx8 may be to facilitate host innate immunity. We show that IL-8, a proinflammatory cytokine and a neutrophil chemoattractant, stimulates the protein expression and apical localization of CAREx8 via activation of AKT/S6K and inhibition of GSK3ß. Apical CAREx8 tethers infiltrating neutrophils at the apical surface of a polarized epithelium. Moreover, neutrophils present on the apical-epithelial surface enhance adenovirus entry into the epithelium. These findings suggest that adenovirus evolved to co-opt an innate immune response pathway that stimulates the expression of its primary receptor, apical CAREx8, to allow the initial infection the intact epithelium. In addition, CAREx8 is a new target for the development of novel therapeutics for both respiratory inflammatory disease and adenoviral infection.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae , Epithelial Cells/metabolism , Immunity, Innate/immunology , Adenoviridae Infections/metabolism , Animals , Cells, Cultured , Epithelium/metabolism , Humans , Mice , Neutrophils/immunology , Receptors, Virus/metabolism
15.
Int J Biochem Cell Biol ; 61: 29-34, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25622559

ABSTRACT

The Coxsackievirus and adenovirus receptor (CAR) is an essential cellular protein that is involved in cell-cell adhesion, protein trafficking, and viral infection. The major isoform of CAR is selectively sorted to the basolateral membrane of polarized epithelial cells where it co-localizes with the cellular scaffolding protein membrane-associated guanylate kinase with inverted domain structure-1 (MAGI-1). Previously, we demonstrated CAR interacts with MAGI-1 through a PDZ-domain dependent interaction. Here, we show that the PDZ3 domain of MAGI-1 is exclusively responsible for the high affinity interaction between the seven exon isoform of CAR and MAGI-1 using yeast-two-hybrid analysis and confirming this interaction biochemically and in cellular lysates by in vitro pull down assay and co-immunoprecipitation. The high affinity interaction between the PDZ3 domain and CAR C-terminus was measured by fluorescence resonance energy transfer. Further, we investigated the biological relevance of this high affinity interaction between CAR and the PDZ3 domain of MAGI-1 and found that it does not alter CAR-mediated adenovirus infection. By contrast, interruption of this high affinity interaction altered the localization of MAGI-1 indicating that CAR is able to traffic MAGI-1 to cell junctions. These data deepen the molecular understanding of the interaction between CAR and MAGI-1 and indicate that although CAR plays a role in trafficking PDZ-based scaffolding proteins to cellular junctions, association with a high affinity intracellular binding partner does not significantly alter adenovirus binding and entry via CAR.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Animals , CHO Cells , COS Cells , Cell Adhesion Molecules, Neuronal/genetics , Cell Membrane/metabolism , Chlorocebus aethiops , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Cricetulus , Models, Molecular , PDZ Domains , Transfection
16.
Recent Res Dev Virol ; 9: 1-24, 2014.
Article in English | MEDLINE | ID: mdl-25621323

ABSTRACT

Understanding the biology of cell surface proteins is important particularly when they are utilized as viral receptors for viral entry. By manipulating the expression of cell surface receptors that have been coopted by viruses, the susceptibility of an individual to virus-induced disease or, alternatively, the effectiveness of viral-based gene therapy can be modified. The most commonly studied vector for gene therapy is adenovirus. The majority of adenovirus types utilize the coxsackievirus and adenovirus receptor (CAR) as a primary receptor to enter cells. Species B adenovirus do not interact with CAR, but instead interact with the cell surface proteins desmoglein-2 (DSG-2) and cluster of differentiation 46 (CD46). These cell surface proteins exhibit varying degrees of alternative mRNA splicing, creating an estimated 20 distinct protein isoforms. It is likely that alternative splice forms have allowed these proteins to optimize their effectiveness in a plethora of niches, including roles as cell adhesion proteins and regulators of the innate immune system. Interestingly, there are soluble isoforms of these viral receptors, which lack the transmembrane domain. These soluble isoforms can potentially bind to the surface of a virus in the extracellular compartment, blocking the ability of the virus to bind to the host cell, reducing viral infectivity. Finally, the diversity of viral receptor isoforms appears to facilitate an assortment of interactions between viral receptor proteins and cytosolic proteins, leading to differential sorting in polarized cells. Using adenoviral receptors as a model system, the purpose of this review is to highlight the role that isoform-specific protein localization plays in the entry of pathogenic viruses from the apical surface of polarized epithelial cells.

17.
PLoS One ; 7(11): e49930, 2012.
Article in English | MEDLINE | ID: mdl-23166798

ABSTRACT

BACKGROUND: Although significant epidemiological evidence indicates that cigarette smoke exposure increases the incidence and severity of viral infection, the molecular mechanisms behind the increased susceptibility of the respiratory tract to viral pathogens are unclear. Adenoviruses are non-enveloped DNA viruses and important causative agents of acute respiratory disease. The Coxsackievirus and adenovirus receptor (CAR) is the primary receptor for many adenoviruses. We hypothesized that cigarette smoke exposure increases epithelial susceptibility to adenovirus infection by increasing the abundance of apical CAR. METHODOLOGY AND FINDINGS: Cultured human airway epithelial cells (CaLu-3) were used as a model to investigate the effect of sidestream cigarette smoke (SSS), mainstream cigarette smoke (MSS), or control air exposure on the susceptibility of polarized respiratory epithelia to adenoviral infection. Using a Cultex air-liquid interface exposure system, we have discovered novel differences in epithelial susceptibility between SSS and MSS exposures. SSS exposure upregulates an eight-exon isoform of CAR and increases adenoviral entry from the apical surface whilst MSS exposure is similar to control air exposure. Additionally, the level of cellular glycogen synthase kinase 3ß (GSK3ß) is downregulated by SSS exposure and treatment with a specific GSK3ß inhibitor recapitulates the effects of SSS exposure on CAR expression and viral infection. CONCLUSIONS: This is the first time that SSS exposure has been shown to directly enhance the susceptibility of a polarized epithelium to infection by a common respiratory viral pathogen. This work provides a novel understanding of the impact of SSS on the burden of respiratory viral infections and may lead to new strategies to alter viral infections. Moreover, since GSK3ß inhibitors are under intense clinical investigation as therapeutics for a diverse range of diseases, studies such as these might provide insight to extend the use of clinically relevant therapeutics and increase the understanding of potential side effects.


Subject(s)
Adenoviridae Infections/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Disease Susceptibility/virology , Gene Expression Regulation/drug effects , Respiratory Mucosa/virology , Tobacco Smoke Pollution/adverse effects , Adenoviridae Infections/etiology , Blotting, Western , Cells, Cultured , DNA Primers/genetics , Electric Impedance , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Immunohistochemistry , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
18.
Biochem Biophys Res Commun ; 425(1): 13-8, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22809504

ABSTRACT

We have previously shown that the Coxsackievirus and adenovirus receptor (CAR) can interact with post-synaptic density 95 (PSD-95) and localize PSD-95 to cell-cell junctions. We have also shown that activity of the acid sensing ion channel (ASIC3), a H(+)-gated cation channel that plays a role in mechanosensation and pain signaling, is negatively modulated by PSD-95 through a PDZ-based interaction. We asked whether CAR and ASIC3 simultaneously interact with PSD-95, and if so, whether co-expression of these proteins alters their cellular distribution and localization. Results indicate that CAR and ASIC3 co-immunoprecipitate only when co-expressed with PSD-95. CAR also brings both PSD-95 and ASIC3 to the junctions of heterologous cells. Moreover, CAR rescues PSD-95-mediated inhibition of ASIC3 currents. These data suggest that, in addition to activity as a viral receptor and adhesion molecule, CAR can play a role in trafficking proteins, including ion channels, in a PDZ-based scaffolding complex.


Subject(s)
Acid Sensing Ion Channels/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Acid Sensing Ion Channels/genetics , Animals , COS Cells , Chlorocebus aethiops , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Disks Large Homolog 4 Protein , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , PDZ Domains , Protein Transport
19.
J Gene Med ; 14(6): 366-73, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22610695

ABSTRACT

BACKGROUND: In airway epithelia, the kinetics of recombinant adeno-associated virus (AAV) transgene expression is slow. This has negative practical implications for research, as well as for translation into therapy. The DNA minor groove-binding agent Hoechst-33342 has been shown to enhance AAV transgene expression. In the present study, we investigated the mechanism of Hoechst-related augmentation of AAV-mediated transgene expression. METHODS: We investigated the effect of Hoechst-33342 on HT1080, COS-7, mouse and human airway epithelia transduced with different AAV serotypes encoding enhanced green fluorescent protein (eGFP). We exposed cells to increasing concentrations of Hoechst-33342 at different time points. We evaluated the effect on second-strand DNA synthesis using AAV with a self-complementary genome. We also investigated the effect on expression from transfected plasmids with and without AAV2 inverted terminal repeats (ITRs). RESULTS: We found that Hoechst-33342 significantly accelerated AAV transgene expression for all serotypes tested. Hoechst-33342 only had an effect when the treatment was given during or after transduction, even 120 days post-transduction, suggesting an effect on transgene expression regulation. Hoechst-33342 increased transgene expression when cells were transduced with a self-complementary AAV with the cytomegalovirus promoter, although there was no effect on cells transduced with conventional single-stranded AAV encoding the Rous sarcoma virus promoter. Finally, Hoechst-33342 increases gene expression from transfected plasmids regardless of the presence of AAV2 ITRs. CONCLUSIONS: Hoechst dramatically augments and accelerates AAV-mediated transgene expression in airway epithelia without altering AAV-mediated gene transfer. Hoechst activation of the cytomegalovirus promoter is seen in plasmids, although it is drastically enhanced in the context of AAV.


Subject(s)
Benzimidazoles/pharmacology , Cytomegalovirus/genetics , Dependovirus/genetics , Gene Expression Regulation , Promoter Regions, Genetic , Respiratory Mucosa/drug effects , Transgenes , Animals , COS Cells , Cell Cycle/drug effects , Cell Line , Chlorocebus aethiops , Cytomegalovirus/drug effects , DNA/drug effects , Dependovirus/drug effects , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Mice , Nucleic Acid Conformation , Plasmids/genetics , Rous sarcoma virus/genetics , Transduction, Genetic
20.
J Virol ; 85(17): 9023-30, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21697483

ABSTRACT

We previously used directed evolution in human airway epithelia to create adeno-associated virus 2.5T (AAV2.5T), a highly infectious chimera of AAV2 and AAV5 with one point mutation (A581T). We hypothesized that the mechanism for its increased infection may be a higher binding affinity to the surface of airway epithelia than its parent AAV5. Here, we show that, like AAV5, AAV2.5T, uses 2,3N-linked sialic acid as its primary receptor; however, AAV2.5T binds to the apical surface of human airway epithelia at higher levels and has more receptors than AAV5. Furthermore, its binding affinity is similar to that of AAV5. An alternative hypothesis is that AAV2.5T interaction with 2,3N-linked sialic acid may instead be required for cellular internalization. Consistent with this, AAV2.5T binds but fails to be internalized by CHO cells that lack surface expression of sialic acid. Moreover, whereas AAV2.5T binds similarly to human (rich in 2,3N-linked sialic acid) and pig airway epithelia (2,6N-linked sialic acid), significantly more virus was internalized by human airway. Subsequent transduction correlated with the level of internalized rather than surface-bound virus. We also found that human airway epithelia internalized significantly more AAV2.5T than AAV5. These data suggest that AAV2.5T has evolved to utilize specific 2,3N-linked sialic acid residues on the surface of airway epithelia that mediate rapid internalization and subsequent infection. Thus, sialic acid serves as not just an attachment factor but is also required for AAV2.5T internalization, possibly representing an important rate-limiting step for other viruses that use sialic acids.


Subject(s)
Dependovirus/pathogenicity , Endocytosis , N-Acetylneuraminic Acid/metabolism , Receptors, Virus/metabolism , Virus Internalization , Animals , Cell Line , Cricetinae , Cricetulus , Epithelial Cells/virology , Humans , Mutation, Missense , Recombination, Genetic , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...