Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Andreas S. Puschnik is active.

Publication


Featured researches published by Andreas S. Puschnik.


Nature | 2016

Genetic dissection of Flaviviridae host factors through genome-scale CRISPR screens

Caleb Marceau; Andreas S. Puschnik; Karim Majzoub; Yaw Shin Ooi; Susan M. Brewer; Gabriele Fuchs; Kavya Swaminathan; Miguel A. Mata; Joshua E. Elias; Peter Sarnow; Jan E. Carette

The Flaviviridae are a family of viruses that cause severe human diseases. For example, dengue virus (DENV) is a rapidly emerging pathogen causing an estimated 100 million symptomatic infections annually worldwide. No approved antivirals are available to date, and clinical trials with a tetravalent dengue vaccine showed disappointingly low protection rates. Hepatitis C virus (HCV) also remains a major medical problem, with 160 million chronically infected patients worldwide and only expensive treatments available. Despite distinct differences in their pathogenesis and modes of transmission, the two viruses share common replication strategies. A detailed understanding of the host functions that determine viral infection is lacking. Here we use a pooled CRISPR genetic screening strategy to comprehensively dissect host factors required for these two highly important Flaviviridae members. For DENV, we identified endoplasmic-reticulum (ER)-associated multi-protein complexes involved in signal sequence recognition, N-linked glycosylation and ER-associated degradation. DENV replication was nearly completely abrogated in cells deficient in the oligosaccharyltransferase (OST) complex. Mechanistic studies pinpointed viral RNA replication and not entry or translation as the crucial step requiring the OST complex. Moreover, we show that viral non-structural proteins bind to the OST complex. The identified ER-associated protein complexes were also important for infection by other mosquito-borne flaviviruses including Zika virus, an emerging pathogen causing severe birth defects. By contrast, the most significant genes identified in the HCV screen were distinct and included viral receptors, RNA-binding proteins and enzymes involved in metabolism. We found an unexpected link between intracellular flavin adenine dinucleotide (FAD) levels and HCV replication. This study shows notable divergence in host-dependency factors between DENV and HCV, and illuminates new host targets for antiviral therapy.


Nature | 2016

An essential receptor for adeno-associated virus infection

Sirika Pillay; Nancy L. Meyer; Andreas S. Puschnik; Omar Davulcu; Jonathan Diep; Yoshihiro Ishikawa; Lucas T. Jae; Jonathan E. Wosen; Claude M. Nagamine; Michael S. Chapman; Jan E. Carette

Adeno-associated virus (AAV) vectors are currently the leading candidates for virus-based gene therapies because of their broad tissue tropism, non-pathogenic nature and low immunogenicity. They have been successfully used in clinical trials to treat hereditary diseases such as haemophilia B (ref. 2), and have been approved for treatment of lipoprotein lipase deficiency in Europe. Considerable efforts have been made to engineer AAV variants with novel and biomedically valuable cell tropisms to allow efficacious systemic administration, yet basic aspects of AAV cellular entry are still poorly understood. In particular, the protein receptor(s) required for AAV entry after cell attachment remains unknown. Here we use an unbiased genetic screen to identify proteins essential for AAV serotype 2 (AAV2) infection in a haploid human cell line. The most significantly enriched gene of the screen encodes a previously uncharacterized type I transmembrane protein, KIAA0319L (denoted hereafter as AAV receptor (AAVR)). We characterize AAVR as a protein capable of rapid endocytosis from the plasma membrane and trafficking to the trans-Golgi network. We show that AAVR directly binds to AAV2 particles, and that anti-AAVR antibodies efficiently block AAV2 infection. Moreover, genetic ablation of AAVR renders a wide range of mammalian cell types highly resistant to AAV2 infection. Notably, AAVR serves as a critical host factor for all tested AAV serotypes. The importance of AAVR for in vivo gene delivery is further highlighted by the robust resistance of Aavr−/− (also known as Au040320−/− and Kiaa0319l−/−) mice to AAV infection. Collectively, our data indicate that AAVR is a universal receptor involved in AAV infection.


Nature Reviews Microbiology | 2017

A CRISPR toolbox to study virus–host interactions

Andreas S. Puschnik; Karim Majzoub; Yaw Shin Ooi; Jan E. Carette

Viruses depend on their hosts to complete their replication cycles; they exploit cellular receptors for entry and hijack cellular functions to replicate their genome, assemble progeny virions and spread. Recently, genome-scale CRISPR–Cas screens have been used to identify host factors that are required for virus replication, including the replication of clinically relevant viruses such as Zika virus, West Nile virus, dengue virus and hepatitis C virus. In this Review, we discuss the technical aspects of genome-scale knockout screens using CRISPR–Cas technology, and we compare these screens with alternative genetic screening technologies. The relative ease of use and reproducibility of CRISPR–Cas make it a powerful tool for probing virus–host interactions and for identifying new antiviral targets.


Journal of Virology | 2017

Adeno-associated Virus (AAV) Serotypes Have Distinctive Interactions with Domains of the Cellular AAV Receptor

Sirika Pillay; Wei Zou; Fang Cheng; Andreas S. Puschnik; Nancy L. Meyer; Safder S. Ganaie; Xuefeng Deng; Jonathan E. Wosen; Omar Davulcu; Ziying Yan; John F. Engelhardt; Kevin E. Brown; Michael S. Chapman; Jianming Qiu; Jan E. Carette

ABSTRACT Adeno-associated virus (AAV) entry is determined by its interactions with specific surface glycans and a proteinaceous receptor(s). Adeno-associated virus receptor (AAVR) (also named KIAA0319L) is an essential cellular receptor required for the transduction of vectors derived from multiple AAV serotypes, including the evolutionarily distant serotypes AAV2 and AAV5. Here, we further biochemically characterize the AAV-AAVR interaction and define the domains within the ectodomain of AAVR that facilitate this interaction. By using a virus overlay assay, it was previously shown that the major AAV2 binding protein in membrane preparations of human cells corresponds to a glycoprotein with a molecular mass of 150 kDa. By establishing a purification procedure, performing further protein separation by two-dimensional electrophoresis, and utilizing mass spectrometry, we now show that this glycoprotein is identical to AAVR. While we find that AAVR is an N-linked glycosylated protein, this glycosylation is not a strict requirement for AAV2 binding or functional transduction. Using a combination of genetic complementation with deletion constructs and virus overlay assays with individual domains, we find that AAV2 functionally interacts predominantly with the second Ig-like polycystic kidney disease (PKD) repeat domain (PKD2) present in the ectodomain of AAVR. In contrast, AAV5 interacts primarily through the first, most membrane-distal, PKD domain (PKD1) of AAVR to promote transduction. Furthermore, other AAV serotypes, including AAV1 and -8, require a combination of PKD1 and PKD2 for optimal transduction. These results suggest that despite their shared dependence on AAVR as a critical entry receptor, different AAV serotypes have evolved distinctive interactions with the same receptor. IMPORTANCE Over the past decade, AAV vectors have emerged as leading gene delivery tools for therapeutic applications and biomedical research. However, fundamental aspects of the AAV life cycle, including how AAV interacts with host cellular factors to facilitate infection, are only partly understood. In particular, AAV receptors contribute significantly to AAV vector transduction efficiency and tropism. The recently identified AAV receptor (AAVR) is a key host receptor for multiple serotypes, including the most studied serotype, AAV2. AAVR binds directly to AAV2 particles and is rate limiting for viral transduction. Defining the AAV-AAVR interface in more detail is important to understand how AAV engages with its cellular receptor and how the receptor facilitates the entry process. Here, we further define AAV-AAVR interactions, genetically and biochemically, and show that different AAV serotypes have discrete interactions with the Ig-like PKD domains of AAVR. These findings reveal an unexpected divergence of AAVR engagement within these parvoviruses.


Cell Reports | 2017

A Small-Molecule Oligosaccharyltransferase Inhibitor with Pan-flaviviral Activity

Andreas S. Puschnik; Caleb Marceau; Yaw Shin Ooi; Karim Majzoub; Natalie Rinis; Joseph N. Contessa; Jan E. Carette

The mosquito-borne flaviviruses include important human pathogens such as dengue, Zika, West Nile, and yellow fever viruses, which pose a serious threat for global health. Recent genetic screens identified endoplasmic reticulum (ER)-membrane multiprotein complexes, including the oligosaccharyltransferase (OST) complex, as critical flavivirus host factors. Here, we show that a chemical modulator of the OST complex termed NGI-1 has promising antiviral activity against flavivirus infections. We demonstrate that NGI-1 blocks viral RNA replication and that antiviral activity does not depend on inhibition of the N-glycosylation function of the OST. Viral mutants adapted to replicate in cells deficient of the OST complex showed resistance to NGI-1 treatment, reinforcing the on-target activity of NGI-1. Lastly, we show that NGI-1 also has strong antiviral activity in primary and disease-relevant cell types. This study provides an example for advancing from the identification of genetic determinants of infection to a host-directed antiviral compound with broad activity against flaviviruses.


Journal of Virology | 2018

An alternate route for adeno-associated virus entry independent of AAVR

Amanda M. Dudek; Sirika Pillay; Andreas S. Puschnik; Claude M. Nagamine; Fang Cheng; Jianming Qiu; Jan E. Carette; Luk H. Vandenberghe

ABSTRACT Determinants and mechanisms of cell attachment and entry steer adeno-associated virus (AAV) in its utility as a gene therapy vector. Thus far, a systematic assessment of how diverse AAV serotypes engage their proteinaceous receptor AAVR (KIAA0319L) to establish transduction has been lacking, despite potential implications for cell and tissue tropism. Here, a large set of human and simian AAVs as well as in silico-reconstructed ancestral AAV capsids were interrogated for AAVR usage. We identified a distinct AAV capsid lineage comprised of AAV4 and AAVrh32.33 that can bind and transduce cells in the absence of AAVR, independent of the multiplicity of infection. Virus overlay assays and rescue experiments in nonpermissive cells demonstrate that these AAVs are unable to bind to or use the AAVR protein for entry. Further evidence for a distinct entry pathway was observed in vivo, as AAVR knockout mice were equally as permissive to transduction by AAVrh32.33 as wild-type mice upon systemic injection. We interestingly observe that some AAV capsids undergo a low level of transduction in the absence of AAVR, both in vitro and in vivo, suggesting that some capsids may have a multimodal entry pathway. In aggregate, our results demonstrate that AAVR usage is conserved among all primate AAVs except for those of the AAV4 lineage, and a non-AAVR pathway may be available to other serotypes. This work furthers our understanding of the entry of AAV, a vector system of broad utility in gene therapy. IMPORTANCE Adeno-associated virus (AAV) is a nonpathogenic virus that is used as a vehicle for gene delivery. Here, we have identified several situations in which transduction is retained in both cell lines and a mouse model in the absence of a previously defined entry receptor, AAVR. Defining the molecular determinants of the infectious pathway of this highly relevant viral vector system can help refine future applications and therapies with this vector.


Journal of Virology | 2017

Monkeypox Virus Host Factor Screen Using Haploid Cells Identifies Essential Role of GARP Complex in Extracellular Virus Formation

Susan Realegeno; Andreas S. Puschnik; Amrita Kumar; Cynthia S. Goldsmith; Jillybeth Burgado; Suryaprakash Sambhara; Victoria A. Olson; Darin S. Carroll; Inger K. Damon; Tetsuya Hirata; Taroh Kinoshita; Jan E. Carette; Panayampalli Subbian Satheshkumar

ABSTRACT Monkeypox virus (MPXV) is a human pathogen that is a member of the Orthopoxvirus genus, which includes Vaccinia virus and Variola virus (the causative agent of smallpox). Human monkeypox is considered an emerging zoonotic infectious disease. To identify host factors required for MPXV infection, we performed a genome-wide insertional mutagenesis screen in human haploid cells. The screen revealed several candidate genes, including those involved in Golgi trafficking, glycosaminoglycan biosynthesis, and glycosylphosphatidylinositol (GPI)-anchor biosynthesis. We validated the role of a set of vacuolar protein sorting (VPS) genes during infection, VPS51 to VPS54 (VPS51–54), which comprise the Golgi-associated retrograde protein (GARP) complex. The GARP complex is a tethering complex involved in retrograde transport of endosomes to the trans-Golgi apparatus. Our data demonstrate that VPS52 and VPS54 were dispensable for mature virion (MV) production but were required for extracellular virus (EV) formation. For comparison, a known antiviral compound, ST-246, was used in our experiments, demonstrating that EV titers in VPS52 and VPS54 knockout (KO) cells were comparable to levels exhibited by ST-246-treated wild-type cells. Confocal microscopy was used to examine actin tail formation, one of the viral egress mechanisms for cell-to-cell dissemination, and revealed an absence of actin tails in VPS52KO- or VPS54KO-infected cells. Further evaluation of these cells by electron microscopy demonstrated a decrease in levels of wrapped viruses (WVs) compared to those seen with the wild-type control. Collectively, our data demonstrate the role of GARP complex genes in double-membrane wrapping of MVs necessary for EV formation, implicating the host endosomal trafficking pathway in orthopoxvirus infection. IMPORTANCE Human monkeypox is an emerging zoonotic infectious disease caused by Monkeypox virus (MPXV). Of the two MPXV clades, the Congo Basin strain is associated with severe disease, increased mortality, and increased human-to-human transmission relative to the West African strain. Monkeypox is endemic in regions of western and central Africa but was introduced into the United States in 2003 from the importation of infected animals. The threat of MPXV and other orthopoxviruses is increasing due to the absence of routine smallpox vaccination leading to a higher proportion of naive populations. In this study, we have identified and validated candidate genes that are required for MPXV infection, specifically, those associated with the Golgi-associated retrograde protein (GARP) complex. Identifying host targets required for infection that prevents extracellular virus formation such as the GARP complex or the retrograde pathway can provide a potential target for antiviral therapy.


Nature | 2016

Corrigendum: An essential receptor for adeno-associated virus infection

Sirika Pillay; Nancy L. Meyer; Andreas S. Puschnik; Omar Davulcu; Jonathan Diep; Yoshihiro Ishikawa; Lucas T. Jae; Jonathan E. Wosen; Claude M. Nagamine; Michael S. Chapman; Jan E. Carette

This corrects the article DOI: 10.1038/nature16465


Molecular Therapy | 2016

478. An Essential and Ubiquitous Protein Receptor for AAV; Glycans as Attachment Receptors

Nancy L. Meyer; Sirika Pillay; Qing Xie; Omar Davulcu; Andreas S. Puschnik; Jonathan Diep; Yoshi Ishikawa; Lucas T. Jae; Jonathan E. Wosen; Claude M. Nagamine; Alex J. Noble; Scott M. Stagg; Jan E. Carette; Michael S. Chapman

Motivated by unsuccessful attempts to observe physical binding between AAV-2 and heterologously expressed domains of previously reported co-receptors, we set out to identify novel protein receptor(s) for AAV2 through an unbiased genome-wide knockout screen in human cells. Using an mCherry AAV vector, resistant cells were iteratively selected by FACS for gene trap screening in a library of mutagenized haploid cells. Upon deep sequencing, refractory cells had significantly high frequencies of mutation in genes encoding glycan synthesis and retrograde transport, but most significantly in a hitherto poorly characterized transmembrane protein, now termed AAVR. Genetic confirmation of AAVRs role in the entry of multiple AAV serotypes has come through CRISPR-Cas9 knockouts in multiple cell lines then restoration of susceptibility through complementation; infection of poorly permissive cells following AAVR transduction; and creation of a mouse knockout with greatly diminished susceptibility.Various AAVR ectodomain constructs have been heterologously expressed and purified as fusion proteins, and these have been shown to inhibit in vitro viral transduction at concentrations consistent with effective nM binding constants (between AAV & AAVR) measured by surface plasmon resonance (SPR). Pre-incubation with antibodies to AAVR also inhibits infection or transduction. AAVR is transiently expressed on the plasma membrane. Expression of chimeric constructs suggests that AAV takes advantage of its trafficking to the perinuclear trans Golgi network as the dominant, but non-exclusive, entry pathway. Identification of AAVR and its apparently ubiquitous use has interesting implications for AAVs cell specificity. Progress towards structure of complexes will be reported.AAVR exhibits the classic characteristics of a viral receptor, casting the roles ascribed to glycan “primary” receptors in new light. Electron microscopy has been used to visualize AAV-DJ in complex with various heparin analogs at increasingly high resolution. A structure at 2.8 A resolution, as a pentasaccharide complex, shows some disorder in the glycan, but the side chains of viral amino acids are clearly resolved and in different conformations from those seen in a sucrose octasulfate complex. With little change to the backbone, the binding site accommodates diverse glycan sequences through adjustments to side chains, consistent with SPR binding assays of AAV-2 to a library of heparanoids. This, together with comparisons of heparan and AAVR cell knock-outs, indicates a more accessory role for glycans than is implied by the term “primary”. As for several other viruses, in AAV-2 at least, the glycan is an attachment receptor that likely elevates the AAV concentration proximal to the membrane, improving the efficiency with which the virus then binds to AAVR.


Molecular Therapy | 2016

293. Discovery of an Essential Receptor for Adeno-Associated Virus Infection

Sirika Pillay; Nancy L. Meyer; Andreas S. Puschnik; Omar Davulco; Jonathan Diep; Yoshi Ishikawa; Lucas T. Jae; Jonathan E. Wosen; Claude M. Nagamine; Michael S. Chapman; Jan E. Carette

Cellular entry of adeno-associated viruses (AAV) is poorly understood, despite the prominent use of AAV vectors in gene therapy for several monogenic diseases. Using an unbiased, haploid genetic screen, we identified critical players in AAV serotype 2 (AAV2) entry including members of distinct protein complexes involved in retrograde trafficking as well as genes involved in the biosynthesis of the AAV2 attachment factor, heparan sulphate. We focused on the single most significantly enriched gene of the screen, an uncharacterized type-I transmembrane protein. Based on the evidence below we renamed this gene AAV receptor (AAVR). We discovered AAVR as capable of rapidly endocytosing from the plasma membrane and trafficking to the trans-Golgi network, taking a similar path as AAV particles utilize. Genetic ablation of AAVR using CRISPR/Cas9 technology demonstrated a robust resistance to AAV2 infection in a wide range of mammalian cell types, which could be reversed upon AAVR complementation. This confirmed the essentiality of AAVR in AAV2 infection. Further investigation revealed that AAVR was also required for the infections of all tested human and simian-derived AAV serotypes including AAV1, 3B, 5, 6, 8 and 9. Deeper characterization of AAVR showed it to contain Ig-like domains, which are commonly found in many virus receptors including those for poliovirus and measles. We observed that these domains are capable of binding to AAV2 particles, and anti-AAVR antibodies efficiently block AAV2 infection. Moreover, the importance of AAVR for AAV infection in vivo was demonstrated by the strong resistance of AAVR−/− mice to AAV9 infection, recapitulating what we showed in vitro. Collectively, the data indicates that AAVR is a universal receptor involved in AAV infection. This has significant implications for the improvement of future AAV vector design.

Collaboration


Dive into the Andreas S. Puschnik's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lucas T. Jae

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge