Network


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

Hotspot


Dive into the research topics where Nancy L. Meyer is active.

Publication


Featured researches published by Nancy L. Meyer.


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.


Virology | 2011

Structure-function analysis of receptor-binding in adeno-associated virus serotype 6 (AAV-6).

Qing Xie; Thomas F. Lerch; Nancy L. Meyer; Michael S. Chapman

Crystal structures of the AAV-6 capsid at 3Å reveal a subunit fold homologous to other parvoviruses with greatest differences in two external loops. The electrostatic potential suggests that receptor-attachment is mediated by four residues: Arg(576), Lys(493), Lys(459) and Lys(531), defining a positively charged region curving up from the valley between adjacent spikes. It overlaps only partially with the receptor-binding site of AAV-2, and the residues endowing the electrostatic character are not homologous. Mutational substitution of each residue decreases heparin affinity, particularly Lys(531) and Lys(459). Neither is conserved among heparin-binding serotypes, indicating that diverse modes of receptor attachment have been selected in different serotypes. Surface topology and charge are also distinct at the shoulder of the spike, where linear epitopes for AAV-2s neutralizing monoclonal antibody A20 come together. Evolutionarily, selection of changed side-chain charge may have offered a conservative means to evade immune neutralization while preserving other essential functionality.


DNA and Cell Biology | 2011

Interaction Between Troponin and Myosin Enhances Contractile Activity of Myosin in Cardiac Muscle

Brenda Schoffstall; Vincent LaBarbera; Nicolas M. Brunet; Belinda J. Gavino; Lauren Herring; Sara Heshmati; Brittany H. Kraft; Vanessa Inchausti; Nancy L. Meyer; Danamarie Moonoo; Aya K. Takeda; Prescott Bryant Chase

Ca(2+) signaling in striated muscle cells is critically dependent upon thin filament proteins tropomyosin (Tm) and troponin (Tn) to regulate mechanical output. Using in vitro measurements of contractility, we demonstrate that even in the absence of actin and Tm, human cardiac Tn (cTn) enhances heavy meromyosin MgATPase activity by up to 2.5-fold in solution. In addition, cTn without Tm significantly increases, or superactivates sliding speed of filamentous actin (F-actin) in skeletal motility assays by at least 12%, depending upon [cTn]. cTn alone enhances skeletal heavy meromyosins MgATPase in a concentration-dependent manner and with sub-micromolar affinity. cTn-mediated increases in myosin ATPase may be the cause of superactivation of maximum Ca(2+)-activated regulated thin filament sliding speed in motility assays relative to unregulated skeletal F-actin. To specifically relate this classical superactivation to cardiac muscle, we demonstrate the same response using motility assays where only cardiac proteins were used, where regulated cardiac thin filament sliding speeds with cardiac myosin are >50% faster than unregulated cardiac F-actin. We additionally demonstrate that the COOH-terminal mobile domain of cTnI is not required for this interaction or functional enhancement of myosin activity. Our results provide strong evidence that the interaction between cTn and myosin is responsible for enhancement of cross-bridge kinetics when myosin binds in the vicinity of Tn on thin filaments. These data imply a novel and functionally significant molecular interaction that may provide new insights into Ca(2+) activation in cardiac muscle cells.


Journal of Virology | 2017

Adeno-associated Virus (AAV) Assembly-Activating Protein Is Not an Essential Requirement for Capsid Assembly of AAV Serotypes 4, 5, and 11

Lauriel F. Earley; John Powers; Kei Adachi; Joshua T. Baumgart; Nancy L. Meyer; Qing Xie; Michael S. Chapman; Hiroyuki Nakai

ABSTRACT Adeno-associated virus (AAV) vectors have made great progress in their use for gene therapy; however, fundamental aspects of AAVs capsid assembly remain poorly characterized. In this regard, the discovery of assembly-activating protein (AAP) sheds new light on this crucial part of AAV biology and vector production. Previous studies have shown that AAP is essential for assembly; however, how its mechanistic roles in assembly might differ among AAV serotypes remains uncharacterized. Here, we show that biological properties of AAPs and capsid assembly processes are surprisingly distinct among AAV serotypes 1 to 12. In the study, we investigated subcellular localizations and assembly-promoting functions of AAP1 to -12 (i.e., AAPs derived from AAV1 to -12, respectively) and examined the AAP dependence of capsid assembly processes of these 12 serotypes using combinatorial approaches that involved immunofluorescence and transmission electron microscopy, barcode-Seq (i. e., a high-throughput quantitative method using DNA barcodes and a next-generation sequencing technology), and quantitative dot blot assays. This study revealed that AAP1 to -12 are all localized in the nucleus with serotype-specific differential patterns of nucleolar association; AAPs and assembled capsids do not necessarily colocalize; AAPs are promiscuous in promoting capsid assembly of other serotypes, with the exception of AAP4, -5, -11, and -12; assembled AAV5, -8, and -9 capsids are excluded from the nucleolus, in contrast to the nucleolar enrichment of assembled AAV2 capsids; and, surprisingly, AAV4, -5, and -11 capsids are not dependent on AAP for assembly. These observations highlight the serotype-dependent heterogeneity of the capsid assembly process and challenge current notions about the role of AAP and the nucleolus in capsid assembly. IMPORTANCE Assembly-activating protein (AAP) is a recently discovered adeno-associated virus (AAV) protein that promotes capsid assembly and provides new opportunities for research in assembly. Previous studies on AAV serotype 2 (AAV2) showed that assembly takes place in the nucleolus and is dependent on AAP and that capsids colocalize with AAP in the nucleolus during the assembly process. However, through the investigation of 12 different AAV serotypes (AAV1 to -12), we find that AAP is not an essential requirement for capsid assembly of AAV4, -5, and -11, and AAP, assembled capsids, and the nucleolus do not colocalize for all the serotypes. In addition, we find that there are both serotype-restricted and serotype-promiscuous AAPs in their assembly roles. These findings challenge widely held beliefs about the importance of the nucleolus and AAP in AAV assembly and show the heterogeneous nature of the assembly process within the AAV family.


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.


Archives of Biochemistry and Biophysics | 2016

Role of cardiac troponin I carboxy terminal mobile domain and linker sequence in regulating cardiac contraction.

Nancy L. Meyer; P. Bryant Chase

Inhibition of striated muscle contraction at resting Ca(2+) depends on the C-terminal half of troponin I (TnI) in thin filaments. Much focus has been on a short inhibitory peptide (Ip) sequence within TnI, but structural studies and identification of disease-associated mutations broadened emphasis to include a larger mobile domain (Md) sequence at the C-terminus of TnI. For Md to function effectively in muscle relaxation, tight mechanical coupling to troponins core-and thus tropomyosin-is presumably needed. We generated recombinant, human cardiac troponins containing one of two TnI constructs: either an 8-amino acid linker between Md and the rest of troponin (cTnILink8), or an Md deletion (cTnI1-163). Motility assays revealed that Ca(2+)-sensitivity of reconstituted thin filament sliding was markedly increased with cTnILink8 (∼0.9 pCa unit leftward shift of speed-pCa relation compared to WT), and increased further when Md was missing entirely (∼1.4 pCa unit shift). Cardiac Tns ability to turn off filament sliding at diastolic Ca(2+) was mostly (61%), but not completely eliminated with cTnI1-163. TnIs Md is required for full inhibition of unloaded filament sliding, although other portions of troponin-presumably including Ip-are also necessary. We also confirm that TnIs Md is not responsible for superactivation of actomyosin cycling by troponin.


Journal of Structural Biology | 2013

Electron microscopy analysis of a disaccharide analog complex reveals receptor interactions of adeno-associated virus.

Qing Xie; Michael Spilman; Nancy L. Meyer; Thomas F. Lerch; Scott M. Stagg; Michael S. Chapman

Mechanistic studies of macromolecular complexes often feature X-ray structures of complexes with bound ligands. The attachment of adeno-associated virus (AAV) to cell surface glycosaminoglycans (GAGs) is an example that has not proven amenable to crystallography, because the binding of GAG analogs disrupts lattice contacts. The interactions of AAV with GAGs are of interest in mediating the cell specificity of AAV-based gene therapy vectors. Previous electron microscopy led to differing conclusions on the exact binding site and the existence of large ligand-induced conformational changes in the virus. Conformational changes are expected during cell entry, but it has remained unclear whether the electron microscopy provided evidence of their induction by GAG-binding. Taking advantage of automated data collection, careful processing and new methods of structure refinement, the structure of AAV-DJ complexed with sucrose octasulfate is determined by electron microscopy difference map analysis to 4.8Å resolution. At this higher resolution, individual sulfate groups are discernible, providing a stereochemical validation of map interpretation, and highlighting interactions with two surface arginines that have been implicated in genetic studies. Conformational changes induced by the SOS are modest and limited to the loop most directly interacting with the ligand. While the resolution attainable will depend on sample order and other factors, there are an increasing number of macromolecular complexes that can be studied by cryo-electron microscopy at resolutions beyond 5Å, for which the approaches used here could be used to characterize the binding of inhibitors and other small molecule effectors when crystallography is not tractable.


Molecular therapy. Methods & clinical development | 2017

The 2.8 Å Electron Microscopy Structure of Adeno-Associated Virus-DJ Bound by a Heparinoid Pentasaccharide

Qing Xie; John M. Spear; Alex J. Noble; Duncan Sousa; Nancy L. Meyer; Omar Davulcu; Fuming Zhang; Robert J. Linhardt; Scott M. Stagg; Michael S. Chapman

Atomic structures of adeno-associated virus (AAV)-DJ, alone and in complex with fondaparinux, have been determined by cryoelectron microscopy at 3 Å resolution. The gene therapy vector, AAV-DJ, is a hybrid of natural serotypes that was previously derived by directed evolution, selecting for hepatocyte entry and resistance to neutralization by human serum. The structure of AAV-DJ differs from that of parental serotypes in two regions where neutralizing antibodies bind, so immune escape appears to have been the primary driver of AAV-DJ’s directed evolution. Fondaparinux is an analog of cell surface heparan sulfate to which several AAVs bind during entry. Fondaparinux interacts with viral arginines at a known heparin binding site, without the large conformational changes whose presence was controversial in low-resolution imaging of AAV2-heparin complexes. The glycan density suggests multi-modal binding that could accommodate sequence variation and multivalent binding along a glycan polymer, consistent with a role in attachment, prior to more specific interactions with a receptor protein mediating entry.


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

12. Assembly-Activating Protein Is Not an Essential Requirement for Capsid Assembly of Adeno-Associated Virus Serotypes 4, 5, and 11

Lauriel F. Earley; John Powers; Kei Adachi; Nancy L. Meyer; Qing Xie; Michael S. Chapman; Hiroyuki Nakai

The mechanisms of capsid assembly for adeno-associated virus (AAV) is still incompletely understood, but delineating the role of the assembly-activating protein (AAP) in this process may lead to insight on this important step in vector production. AAP is a non-structural protein encoded within the cap gene in an overlapping open reading frame (ORF) that initiates near the N-terminus of the VP2 ORF and continues into the VP3 ORF. This protein has been shown to be essential for assembly of AAV2, 8, and 9 capsids; however, interestingly, we have recently found that AAV5 is capable of forming VP3-only capsids without AAP, raising a new question of what role AAP plays in the AAV5 virus life cycle. To further understand the functional role of AAP of AAV5 and other serotypes, it is important to investigate whether AAV5 would require AAP to form infectious capsids containing all three capsid proteins (VP1, VP2, and VP3) and whether other AAV serotypes exist that could form AAP-independent capsids. Here we show that AAV5 particles assembled in the absence of AAP are infectious and AAP-independent capsid assembly takes place for AAV4 and 11 as well but not for AAV12, a serotype that is phylogenetically closely related to AAV4 and 11. In the study, we created an AAV5 helper plasmid expressing AAV2 Rep and AAV5 Cap in which the AAP5 ORF was codon-modified extensively to prevent AAP expression while leaving the VP ORF intact. Using this AAP-KO helper plasmid and the standard AAV5 helper plasmid with an intact AAP ORF, we produced two types of AAV5 vectors expressing a marker gene, AAV5(AAP-) and AAV5(AAP+) vectors, respectively. These vectors were applied on CHO cells, and the marker gene expression was quantified 2 days post-infection. This assay revealed that the AAP-independent AAV5(AAP-) vector was able to efficiently transduce CHO cells although the transduction efficiency was about a half of that with the AAV5(AAP+) vector. The yield of AAV5(AAP-) vector production was lower than AAV5(AAP+) vector by ~10 fold but restored by co-expression of AAP in trans, indicating that AAP retains the assembly promoting role even though AAP is not an absolute requirement for AAV5 capsid assembly. We next sought to determine if AAV4 and 11 could also assemble capsids without AAP. To address this question, we transfected HEK293 cells with codon-modified AAV4 or AAV11 VP3-expressing plasmids and purified the potential viral particles by cesium-chloride ultracentrifugation. Electron microscopy revealed viral particles consistent with AAV capsids. Given the close evolutionary relationship between AAV4, 11 and 12, we hypothesized that AAV12 would also be capable of AAP-independent capsid formation, but preliminary evidence has shown this hypothesis to be incorrect. We anticipate that closer examination of the dependence or independence of viral capsid assembly on AAP among various serotypes will reveal differences in VP structure or the assembly process that will illuminate the mechanistic function of AAP.

Collaboration


Dive into the Nancy L. Meyer'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge